WO2008056779A1 - Procédé destiné à la culture et au passage d'une cellule souche embryonnaire de primate, et procédé destiné à induire la différenciation de la cellule souche embryonnaire - Google Patents

Procédé destiné à la culture et au passage d'une cellule souche embryonnaire de primate, et procédé destiné à induire la différenciation de la cellule souche embryonnaire Download PDF

Info

Publication number
WO2008056779A1
WO2008056779A1 PCT/JP2007/071811 JP2007071811W WO2008056779A1 WO 2008056779 A1 WO2008056779 A1 WO 2008056779A1 JP 2007071811 W JP2007071811 W JP 2007071811W WO 2008056779 A1 WO2008056779 A1 WO 2008056779A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
embryonic stem
culture
blood
Prior art date
Application number
PCT/JP2007/071811
Other languages
English (en)
Japanese (ja)
Inventor
Akira Yuo
Kumiko Tobe
Koichi Saeki
Masako Nakahara
Naoko Nakamura
Yoshiko Yogisashi
Satoko Matsuyama
Asako Yoneda
Original Assignee
Japan As Represented By The President Of International Medical Center Of Japan
Mitsubishi Tanabe Pharma Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Japan As Represented By The President Of International Medical Center Of Japan, Mitsubishi Tanabe Pharma Corporation filed Critical Japan As Represented By The President Of International Medical Center Of Japan
Priority to US12/514,207 priority Critical patent/US20110151554A1/en
Priority to JP2008543145A priority patent/JP5067949B2/ja
Priority to EP07831542A priority patent/EP2088190A4/fr
Publication of WO2008056779A1 publication Critical patent/WO2008056779A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/70Undefined extracts
    • C12N2500/80Undefined extracts from animals
    • C12N2500/84Undefined extracts from animals from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/105Insulin-like growth factors [IGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/125Stem cell factor [SCF], c-kit ligand [KL]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/165Vascular endothelial growth factor [VEGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/22Colony stimulating factors (G-CSF, GM-CSF)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/26Flt-3 ligand (CD135L, flk-2 ligand)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/90Substrates of biological origin, e.g. extracellular matrix, decellularised tissue

Definitions

  • the present invention relates to a method for culturing and subculturing primate embryonic stem cells in an undifferentiated state, and inducing differentiation of the cells into various cells such as blood cells and vascular endothelial precursor cells.
  • the present invention relates to a method, a method for expanding and regenerating the obtained cells, blood cells, vascular endothelial progenitor cells and the like.
  • Embryonic stem cells as universal cells that can be differentiated into various cells were established in the 1980s in mice, and their culture and passage methods are known. However, there has not yet been established a method for continuously culturing and substituting embryonic stem cells derived from primates including humans in an undifferentiated state.
  • Human embryonic stem cells are extremely important in research in the biological and medical fields, and in the clinical field. In particular, a stable supply is required as a basic material for organ preparation in the fields of regenerative medicine and transplantation medicine.
  • organs and tissues created from human embryonic stem cells are expected not only to be used in transplantation and regenerative medicine, but also to enable preclinical studies using human organs that have been limited in practice. .
  • various organs can be produced from human embryonic stem cells, it will be possible to estimate the effects of drugs on the fetus by conducting pharmacological tests during the production process! It has been.
  • Patent Document 1 discloses an embryonic stem cell that is essentially free of feeder cells and is obtained by culturing primate embryonic stem cells using an extracellular matrix prepared from mouse embryonic fibroblasts (MEF). Containing cultures are described.
  • the above established human embryonic stem cells induce chromosomal aberrations after a short passage (specifically, accumulation of chromosomal abnormalities is confirmed after more than a dozen passages). The risk in clinical application has been pointed out (Non-patent Document 1)).
  • Non-patent Document 3 human embryonic stem cell lines were established in Australia and Sweden, and it has been reported that chromosomal abnormalities are extremely rare even after one year or more after establishment. These human embryonic stem cells have the ability to be maintained without feeders S, special culture equipment and special culture skills are required for culturing, subculturing, and freezing, and handling of large amounts of cells is practical It is difficult (Non-Patent Document 3). In addition, feeder-free culturing of these heterologous animal-derived cells without using a conditioned medium requires the use of a large amount of synthetic site force-in, which places a great economic burden on the research itself. Therefore, with regard to human embryonic stem cells, a stable feeder-free culture technique has been established!
  • a method for culturing and passaging primate embryonic stem cells including humans without using a conditioned medium of a heterologous animal-derived cell, a feeder cell, and a synthetic site force-in.
  • a simple basic culture technique allows safe passage without causing chromosomal abnormalities for long periods of time.
  • Such technologies are expected to contribute greatly to the promotion of extensive research, clinical medicine related to transplantation and regeneration, and the development of perinatal medicine.
  • the desired progenitor cells and mature cells can be produced in large quantities and used for various purposes.
  • differentiation of embryonic stem cells can be effectively induced.
  • the method of subculture has been studied.
  • One of the differentiation lines of embryonic stem cells is differentiation into blood-related cells such as blood vessels and blood cells (see Figure 11).
  • Vascular endothelial cells are the basic elements that make up blood vessels. Blood vessels are distributed in almost all living tissues except for some tissues such as cartilage and sclera, and play an extremely important role in supplying nutrients and removing waste products. Therefore, vascular endothelial cells are useful for angiogenesis therapy in obstructive vascular disorders associated with arteriosclerosis, which is a disease of life-style related diseases that have been increasing in recent years. In addition, it is known that the direction of vascular endothelial cells is important for the movement of tissue-specific stem cells (such as neural stem cells) to the appropriate position in the regeneration process of various tissues including the brain. .
  • tissue-specific stem cells such as neural stem cells
  • vascular endothelial cells have played an important role in blood cell production since the embryonic period as vascular endothelial cells function as precursors of blood cells, and in adults, they also play an important role as a hematopoietic stem cell sachet (holding place). Is responsible.
  • vascular endothelial cells are extremely important not only as vascular components but also in general tissue regeneration, including nerves and blood cells. When considering regenerative medicine in general, the control of vascular endothelial cell production is important. It is a problem.
  • vascular endothelial progenitor cells obtained from biological tissues has been attempted S.
  • An example of directly confirming regeneration of vascular endothelial cells from transplanted cells is Absent.
  • mature vascular endothelial cells obtained from living organisms have already lost their proliferative capacity, and basic research on vascular endothelial cells has been extremely delayed in primates where it is difficult to prepare large specimens.
  • primary cultured vascular endothelial cells obtained from a human body especially commercially available primary cultured vascular endothelial cells that have been frozen and thawed have inherent properties in vivo.
  • the differentiation efficiency from mouse embryonic stem cells to vascular endothelial progenitor cells is 90% or more, whereas primate embryonic stem cells differentiate into vascular endothelial progenitor cells.
  • the efficiency is extremely low at 2% or less (Non-Patent Document 4 and Non-Patent Document 5).
  • a method for producing vascular endothelial progenitor cells that can be subcultured in feeder-free culture not only for primate embryonic stem cells but also for mouse embryonic stem cells has not been established.
  • vascular endothelial progenitor cells and mature vascular endothelial cells that can be stably subcultured and freeze-thawed in a feeder-free culture are efficiently produced and cultured from primate embryonic stem cells.
  • Establishing high-volume production technology is necessary for the advancement of regenerative transplant technology, basic medical research on blood vessels, and the development of new pharmacological / toxicological studies that will be effective tools for preclinical trials. Has become an urgent need worldwide.
  • the development of such technology is extremely useful in the medical industry such as basic medical research, clinical medical development and drug discovery.
  • blood (blood cell) cells play an important role in the immune system and are resistant to foreign substances that enter the body, effects on cancer (NK cells), effects on leukemia (hematopoietic stem cells, etc.) Etc.
  • NK cells effects on cancer
  • leukemia hematopoietic stem cells, etc.
  • hematopoietic stem cells are capable of transdifferentiation into cells required for various diseases because of their tissue plasticity, and leukocyte progenitor cells The importance of medical treatment is enormous, for example, by promoting tissue regeneration through fusion.
  • hematopoietic stem cells bone marrow blood, umbilical cord blood, etc.
  • the amount used in transplantation medicine is limited (specimen from one donor). Can only be administered to a single patient) and is virtually impossible to use in basic medical research, including culture experiments.
  • basic research is much slower in primates where it is difficult to prepare large amounts of specimens than in experimental animals such as mice. [0013] Therefore, in addition to application to regenerative medicine, disease treatment, etc., in terms of research, blood cells and hematopoietic stem cells are produced safely and efficiently from human embryonic stem cells by feeder-free culture. There is a long-awaited way to maintain the passage.
  • Blood cells such as erythrocytes thus obtained are useful for improving the safety of treatment free from contamination by AIDS, hepatitis C virus, and the like. Furthermore, transfusion of leukocytes including neutrophils derived from embryonic stem cells can be used to solve problems such as nosocomial infections in order to strengthen the immune system whose function has been reduced by chemotherapy or the like in cancer. In addition, since blood cells lead to an increase in natural healing power, the production of blood cells from embryonic stem cells is thought to provide tremendous benefits for medical care.
  • hematopoietic stem cells can be used for transplantation as well as for the production of blood cells.
  • the efficiency of inducing differentiation from primate embryonic stem cells including humans into blood cells in a feeder-free environment is sufficiently high, and the amount of blood cells produced is also limited.
  • the production efficiency of hematopoietic stem cells is very low (the production efficiency of hematopoietic stem cells is about 5%) (Non-patent Document 6).
  • Non-patent Document 6 Non-patent Document 6
  • stromal cells are indispensable for hematopoiesis in the living body, and the function of hematopoietic stromal cells obtained from living bodies (fetal liver, adult bone marrow, etc.) is rapidly lost by passage or freeze-thawing. Due to the problems, the use in transplantation / regenerative medicine is very limited and in vitro culture is virtually impossible. Therefore, it is essential to establish a technique for producing hematopoietic stromal cells from embryonic stem cells in order to develop transplantation medicine in blood diseases and basic medical research on hematopoietic stromal cells. However, the production of hematopoietic stromal cells from all embryonic stem cells, including mice! /, Has not been successful.
  • the above blood-related cells are useful for elucidating the "hematopoietic mechanism" in primates including humans.
  • hematopoietic mechanisms especially early hematopoiesis in development.
  • common progenitor cells hemangioblasts
  • vascular endothelial cells and blood cells which has been suggested for a long time in the developmental process of mice, has become very recently (September 2006).
  • Zebrafish has only been demonstrated (Non-Patent Document 7). Understanding the hematopoietic function once the technology to produce this “common progenitor cell” from human embryonic stem cells has the only potential in humans to correctly mimic early hematopoiesis in vivo It will be useful for research and medical development.
  • blood-related cells manufactured from primate embryonic stem cells including humans
  • blood-related cells that are capable of stable passage maintenance, expansion reproduction, and freeze-thawing in a feeder-free culture with high efficiency.
  • Blood stem cells mature blood cells, vascular endothelial cells and “common progenitor cells of blood cells”, vascular endothelial cells, hematopoietic stroma cells, etc. It is widely demanded for the development of new pharmacological / toxicity studies that are effective means of preclinical trials.
  • Patent Document 1 International Publication No. 99/20741 Pamphlet
  • Non-Patent Document 1 Draper et al., Nature Biotechnology, Vol. 22, p. 53, p. 54, (2004)
  • Non-Patent Document 2 Ludwig et al., Nature Biotechnology, Vol. 24, 185, 187, (2006)
  • Non-Patent Document 3 Buzzard, Nature Biotechnology, Vol. 22, p. 381, p. 382, (2004)
  • Non-Patent Document 4 Sone et al., Circulation, Vol. 107, p. 2085, p. 2088, (2003)
  • Non-Patent Document 5 Levenberg, Proceeding of National Academy of Science, USA 99, 4391, 4396, (2002)
  • Non-Patent Document 6 Chadwick et al., Blood, Vol. 102, 906, 915, ((22000033))
  • Patent Literature Reference 77 VVooggeellii et al., NNaattuurree, Vol. 444433, pp. 333377, 333399, ((22000066)) My disclosure
  • This invention invents varieties such as primate primate animal and animal embryo embryonic stem stem cell vesicles, chromosomal abnormalities, etc. This is the place where you can provoke the safety and safety of your body. The purpose of this is to provide and provide a method of scrutiny. .
  • the present invention remains unaffected, the primate animal animal embryo embryonic stem stem cell vesicles are efficiently and safely fed into the blood vessel endothelium endothelial cell cysts, Intravascular preendodermis progenitor cell cell, blood blood fluid cell cell, bone marrow medullary cell cell cell, hematopoietic hematopoietic stem cell cell, etc.
  • This place which provides and provides the method of how to skate, is considered to be a white and white fox. .
  • the present invention is related to the blood vessel endothelium endothelial cell cell obtained by the method described above, blood blood fluid cell cell, bone marrow For the purpose of providing and providing medullary cells, hematopoietic hematopoietic stem cells, etc. ! // .
  • the inventors of the present invention have developed a sufficient number of primate animal and animal embryonic stem stem cell vesicles suitable for clinical clinical use. In addition to applying the appropriate amount, various kinds of fine cell vesicles, organ organs, etc.
  • primate animals and embryos embryonic stem stem cell vesicles continue to be maintained in a stable and stable manner. To find out what is held here and where it is guided to the induction of differentiation into various cell vesicles in an appropriate and appropriate manner. As a result, the present invention has been completed. .
  • the gist of the present invention includes the following. .
  • (AA) Primate primate animal embryo embryonic stem stem cell vesicles in a container that has been co-coated with extra-cellular extracellular mammary trix cox. Steps for culturing and cultivating in a culture medium containing protein protein components under a non-fidelity feeder and a non-satisfaction force,
  • step ((CC)) The heart of the embryonic stem stem cell vesicle obtained in the above step ((BB)) A culture medium containing a protein protein component in a container that has been co-coated with a non-fidelity and non-satisfaction force.
  • step (A) The culture and subculture method according to [1] above, wherein the primate embryonic stem cell colony is cultured in step (A) until the colony size is about 2 to about 4 times.
  • step (B) The culture and subculture according to any one of [1] to [3] above, wherein the cell detachment agent in step (B) is at least one selected from the group consisting of trypsin, collagenase, and dispase. Method.
  • step (A) Human collagen, human laminin, human vitronectin, human fibronectin and human serum, and a degradation product thereof, and one kind selected from the group consisting of these synthetic peptides, The culture and subculture method according to any one of [1] to [4].
  • step (B) Step for producing specific progenitor cells including floating cells and adherent cells by adhesion culture of the embryoid body or embryoid body-like cell aggregate obtained in step (A) in the presence of site force-in.
  • step (C) A method for producing blood cells and / or vascular endothelial progenitor cells from primate embryonic stem cells, comprising a step of separating floating cells and adherent cells from the specific progenitor cells obtained in step (B).
  • step (A) The culturing of step (A) is carried out until an embryoid body-like cell aggregate is formed.
  • Cytotensity levels are vascular endothelial growth factor (VEGF), bone morphogenetic protein 4 (BMP4), stem cell factor (SCF), Flt3—ligand (FU, interleukin 6 (IL6), interleukin 3 (IL3), It consists of granulocyte colony stimulating factor (G—CSF), megakaryocyte growth factor (TPO), oncostatin M (OSM), fibroblast growth factor 2 (FGF2) and granulocyte macrophage colony stimulating factor (GM—CSF). At least one selected from the group, 6.
  • a method for producing vascular endothelial progenitor cells and / or blood cells from primate embryonic stem cells according to [7].
  • step (C) The primate according to any one of [6] to [8] above, wherein a cell detachment agent is used in step (C) to separate adherent cells of specific cell precursor cells.
  • a cell detachment agent is used in step (C) to separate adherent cells of specific cell precursor cells.
  • step (B) a step of producing specific precursor cells including floating cells and adherent cells by adhesion culture of the embryoid body or embryoid body-like cell aggregate obtained in step (A) in the presence of site force-in, and
  • step (C) a blood cell, a myeloid cell, a hematopoietic stroma cell and / or a primate embryonic stem cell comprising a step of culturing the specific progenitor cell obtained in step (B) while separating floating cells.
  • a method for producing hematopoietic stem cells comprising a step of culturing the specific progenitor cell obtained in step (B) while separating floating cells.
  • step (A) The culture in step (A) is carried out until the formation of embryoid bodies, blood cells, myeloid cells, hematopoietic stroma cells and / or primate embryonic stem cells according to [11] above.
  • a method for producing hematopoietic stem cells is carried out until the formation of embryoid bodies, blood cells, myeloid cells, hematopoietic stroma cells and / or primate embryonic stem cells according to [11] above.
  • Cytotensity is vascular endothelial growth factor (VEGF), bone morphogenetic protein 4 (BMP4), stem cell factor (SCF), Flt3—ligand (FU, interleukin 6 (IL6), interleukin 3 (IL3) , Granulocyte colony stimulating factor (G—CSF), megakaryocyte growth factor (TPO), oncostatin M (OSM), fibroblast growth factor 2 (FGF2) and granulocyte macrophage colony stimulating factor (GM—CSF)
  • VEGF vascular endothelial growth factor
  • BMP4 stem cell factor
  • SCF stem cell factor
  • Flt3—ligand FU
  • IL6 interleukin 6
  • IL3 interleukin 3
  • G—CSF Granulocyte colony stimulating factor
  • TPO megakaryocyte growth factor
  • OSM oncostatin M
  • FGF2 fibroblast growth factor 2
  • GM—CSF granulocyte macrophage colony stimulating factor
  • step (C) cell detachment is performed to separate adherent cells of specific progenitor cells.
  • a substantially isolated vascular endothelial progenitor cell which is induced to differentiate from a primate embryonic stem cell by the production method according to any one of [6] to [; 10].
  • [17] A substantially isolated blood cell that is induced to differentiate from a primate embryonic stem cell by the production method according to any one of [6] to [; 14].
  • a substantially isolated hematopoietic stem cell which is induced to differentiate from a primate embryonic stem cell by the production method according to [1], [11] to [;
  • a substantially isolated myeloid cell which is induced to differentiate from a primate embryonic stem cell by the production method according to any one of [11] to [; 14].
  • a composition comprising substantially isolated vascular endothelial progenitor cells, blood cells, hematopoietic stroma cells or hematopoietic stem cells according to any one of [16] to [; 19].
  • a primate embryonic stem cell can be safely put in an undifferentiated state by a simple apparatus and method that does not cause cell damage such as chromosomal aberration. It can be maintained and cultured. Further, according to the method of the present invention, primate embryonic stem cells can be maintained in an undifferentiated manner at low cost, and it is possible to widely meet demands in regenerative medicine and research fields.
  • specific cell precursor cells as “common precursor cells” such as vascular endothelial cells and blood cells can be produced safely and with high efficiency.
  • vascular endothelial cells blood cells, hematopoietic stem cells, myeloid cells and the like that can be stably subcultured and frozen and thawed with high reproductivity.
  • a highly safe blood product for blood transfusion including hematopoietic stem cell transplantation, granulocyte transfusion, myeloid cell administration, etc.
  • a material for treatment of vascular injury and improvement of local blood flow is possible to easily supply various cells as materials suitable for the purpose of clinical use for the purpose of promoting the regeneration of various other tissues!
  • the present invention it is possible for the first time to provide a large number of cell groups having a property of correctly mimicking a primate, particularly a human biological tissue. Since these cells can be used appropriately in drug effect assessment tests and toxicity tests, they can greatly contribute to the development of the medical industry as well as clinical medicine.
  • FIG. 1 shows a culture coated with a Matrigel (registered trademark) matrix according to the “undifferentiated maintenance passage method under feeder-free and site-free force-in” of the present invention shown in Example 1. It is a photograph which shows the colony of the power two cynomolgus embryonic stem cell cultured using the dish. A shows cells that were passaged 20 and B was frozen at thirty-five passages and thawed one more time after thawing. The scale bar indicates 100.
  • Matrigel registered trademark
  • Fig. 2 shows a culture coated with a Matrigel (registered trademark) matrix according to the "method of maintaining undifferentiation under feeder-free and site-free force-in" according to the present invention shown in Example 1.
  • This is a result of measuring the expression of SSEA-4 and Oct-4, which are undifferentiation maintenance markers, by flow cytometry in the 20th-generation force cynomolgus embryonic stem cells cultured in a dish. In both cases, very high! /, Expression (> 95%) is confirmed.
  • Fig. 3 shows a culture coated with a Matrigel (registered trademark) matrix according to the "method of maintaining undifferentiation under feeder-free and site-free force-in" of the present invention shown in Example 1. It is the photograph which showed the expression of Tra-1_60, Tra-l_81, and Nanog which is an undifferentiation maintenance marker by the immuno-staining in the 20th-passage force cynomolgus embryonic stem cell culture
  • the scale bar indicates 1 00 ⁇ m.
  • Fig. 4 shows a culture coated with a Matrigel (registered trademark) matrix according to the "undifferentiated maintenance passaging method under feeder-free and site-free force-in" of the present invention shown in Example 1.
  • 3 immunodeficient mice with 21-second-powered cynomolgus embryonic stem cells cultured in a dish It is a photograph of the testis two months after transplantation under the testicular membrane of (SCID mouse). As shown in FIG. 4, tumor formation was confirmed in all three animals.
  • Fig. 5 is a tissue sample of the above tumor (hematoxylin 'eosin staining). Neural epithelium, teeth, secretory gland, intestinal tract-like epithelium, smooth muscle, etc. are observed as described.
  • Fig. 6 shows a culture coated with a Matrigel (registered trademark) matrix by the "undifferentiated maintenance passaging method under feeder-free and site-free force-in" of the present invention shown in Example 2.
  • a phase-contrast photomicrograph of a colony of human embryonic stem cells of passage 24 cultured using a dish is shown.
  • the scale bar indicates 100 ⁇ u m.
  • Fig. 7 shows a culture coated with a Matrigel (registered trademark) matrix according to the "method of maintaining undifferentiation under feeder-free and site-free force-in" of the present invention shown in Example 2.
  • This is a result of measuring the expression of SSEA-4 and Oct-4, which are undifferentiation maintenance markers, in cultured human embryonic stem cells in passage 20 using flow dishes by flow cytometry. In both cases, a very high level and expression (> 95%) were confirmed.
  • Fig. 8 shows a culture coated with a Matrigel (registered trademark) matrix according to the "undifferentiated maintenance passage method under feeder-free and site-free force-in" of the present invention shown in Example 2. It is the photograph which showed the expression of the undifferentiation maintenance marker Oct-4 (A) and Nanog (B) in the human embryonic stem cell of the 25th passage cultured using the dish by the immuno-staining. It is confirmed that both proteins are expressed in almost all cells.
  • the scale bar indicates 100 ⁇ .
  • FIG. 9 is a chromosome analysis diagram (G band method) of human embryonic stem cells.
  • the left is maintained with the conventional culture method recommended by the establishment organization (co-culture method using fetal mouse fountain fibroblasts as one feeder cell)! This is the result of the 20th passage in the “Culture method using site-free force-in”. It was confirmed that no chromosomal abnormality occurred.
  • FIG. 10A shows human-derived fibronectin (5 H g / cm 2 ) according to the “method of maintaining undifferentiation under feeder-free and site-free force-in” of the present invention shown in Example 3.
  • 4 is a phase contrast micrograph of human embryonic stem cells in passage 4 cultured using a culture dish coated only with Resolve the ability to hold undifferentiated forms.
  • FIG. 10B shows the results of measuring the expression of SSEA-4 and Oct-4, which are undifferentiation maintenance markers, in the cells of FIG. 10A by flow cytometry. High expression of both markers was confirmed.
  • FIG. 10C shows a culture dish coated only with human type AB serum according to the “non-differentiation maintenance passaging method under feeder-free and site-free force-in” of the present invention shown in Example 3.
  • Fig. 5 is a phase contrast micrograph of human embryonic stem cells cultured at passage 4 after culturing V. Resolve the ability to hold undifferentiated forms.
  • FIG. 10D shows the results of measuring the expression of SSEA-4 and Oct-4, which are undifferentiation maintenance markers, in the cells of FIG. 10C by flow cytometry. High expression of both markers was confirmed.
  • FIG. 11 shows a differentiation lineage diagram from hematopoietic stem cells to blood cells.
  • FIG. 12 shows the presence of fetal bovine serum from forceped cynomolgus embryonic stem cells by the technique of inducing differentiation and expanding reproduction of vascular endothelial cells' blood cells using a feeder-free feeder of the present invention shown in Example 4.
  • the “specific progenitor cells” structures consisting of sac-like structures and spherical cell populations) that are common to vascular endothelial progenitor cells and blood cells are shown.
  • the scale bar indicates 100 ⁇ m.
  • Fig. 13 shows Wright-Giemsa staining (A) and special staining (myoperoxidase staining (B) and esterase) of mature blood cells produced from spherical cells among the above "specific progenitor cells”. Double staining (C)) is shown. Various myeloid cells are observed, ie cells at each stage of differentiation ranging from myeloblasts to mature blood cells (neutrophils and macrophages). The scale bar indicates 20 m.
  • FIG. 14 shows the expression of VE-cadherin, a vascular endothelial cell-specific marker, and N-cadherin, which is one of vascular endothelial cell markers, of vascular endothelial cells produced from the above-mentioned “specific progenitor cells”.
  • VE-cadherin a vascular endothelial cell-specific marker
  • N-cadherin which is one of vascular endothelial cell markers, of vascular endothelial cells produced from the above-mentioned “specific progenitor cells”.
  • the scale bar indicates 50 m.
  • FIG. 15 shows the expression of PECAM1, which is a marker of mature vascular endothelium, in vascular endothelial cells produced from the above-mentioned “specific progenitor cells” and VE, which is a vascular endothelium-specific marker. This was confirmed by flow cytometry by double staining with -cadherin.
  • the horizontal axis represents PECAM1, and the vertical axis represents the expression intensity of VE-cadherin. 40% or more In both cells, the expression of both is confirmed.
  • FIG. 16 shows the code-forming ability (A) and acetylated low-density lipoprotein (AC-LDL) for confirming the maturation function of vascular endothelial cells produced from the above-mentioned “specific progenitor cells”. This is an investigation of uptake capacity (B). V, it ’s too high! / Is very high!
  • FIG. 17 is a serum-free culture from a forceless cynomolgus embryonic stem cell according to the “method for inducing differentiation into vascular endothelial cells and blood cells without a feeder” of the present invention shown in Example 4. Shows Wright-Giems a staining (A) and special staining (myelin peroxidase staining (B) and esterase double staining (C)) of mature blood cells prepared under conditions (using KNOCKOUT (registered trademark) SR) . Various myeloid cells are observed, ie cells at each stage of differentiation ranging from myeloblasts to mature blood cells (neutrophils and macrophages). The scale bar indicates 20.
  • FIG. 18 shows the serum-free condition from a forceless cynomolgus embryonic stem cell according to the “method for inducing differentiation into vascular endothelial cells and blood cells without a feeder” of the present invention shown in Example 5.
  • the expression of VE-cadherin and PECAM1 in the vascular endothelial cells prepared in (using K NOCKOUT (registered trademark) SR) was confirmed by flow cytometry.
  • K NOCKOUT registered trademark
  • PECAM1 cell membrane expression was confirmed with an efficiency of more than several percent, which was believed to be the inability to induce differentiation of endodermal cells without serum.
  • FIG. 19 is a graph showing fetal bovine serum obtained from force-fed cynomolgus embryonic stem cells according to the “method for inducing differentiation into vascular endothelial cells and blood cells without feeder” shown in Example 6.
  • the scale bar indicates 100.
  • Fig. 20 is a phase-contrast photomicrograph showing the situation in which expanded reproduction of blood cells is performed by culturing the above-mentioned “specific progenitor cells”. Both hematopoietic stroma cells (adherent cells) and blood cells produced from them (floating cells) are confirmed.
  • the scale bar is 10 0 u m.
  • Fig. 21A-D shows the Wright-Giemsa stained image (A) of the blood cells (floating cells) collected during the process of expanding and regenerating the blood cells described above, and special staining (Mie mouth peroxidase staining). (B), esterase double staining (C), neutrophilic alkaline phosphatase staining (D)) Indicates. Various myeloid cells are observed, ie cells at each stage of differentiation ranging from myeloblasts to mature blood cells (neutrophils and macrophages). The scale bar indicates 20.
  • FIG. 21E shows the results of confirming the expression of CD34, which is one hematopoietic stem cell marker, in the blood cells (floating cells) of FIGS. 21A-D by flow cytometry.
  • FIG. 21F shows the results of confirming the expression of CD45 (F), which is a pan-blood cell marker, in the blood cells (floating cells) in FIGS. 21A-D by flow cytometry.
  • FIG. 22A is a phase contrast micrograph (a scale bar indicates 100 m) of a “passable hematopoietic stroma cell” produced from the above-mentioned “specific progenitor cells”.
  • FIG. 22B shows the results of the flow cytometry of the expression of CD34 and CD45 of the passable “hematopoietic stroma cells” produced from the above-mentioned “specific progenitor cells”.
  • Both the pancytopenic marker CD45 and the hematopoietic stem cell marker CD34 are almost negative (small positive cells are considered to be contaminated with hematopoietic stem cells adhering to hematopoietic stroma cells).
  • Fig. 23A shows “CD34-positive and CD45-positive” produced by long-term culture (> 100 days) of “passable hematopoietic stem / progenitor cells” produced from the above-mentioned “specific progenitor cells”. It is a phase-contrast micrograph of “cells”. Although floating cells and adherent cells coexist, they can migrate to each other and are considered to be equivalent cell populations (ie, hematopoietic stem cells and equivalents). The scale bar indicates 100.
  • Fig. 23B shows “CD34-positive and CD45-positive” produced by long-term culture (> 100 days) of “passable hematopoietic stem / progenitor cells” produced from the above-mentioned “specific progenitor cells”. The results of confirming the expression of CD34 and CD45 with respect to the floating cells and adherent cells in “cells” by flow cytometry are shown.
  • FIG. 24 is a phase-contrast micrograph of the situation in which the suspension was frozen and thawed and the culture was resumed in the situation where the above-mentioned “expanded reproduction of blood cells” was performed. In all cases, the presence of hematopoietic stromal cells (adherent cells) and blood cells (floating cells) produced therefrom were confirmed just as before freezing. The scale bar indicates 100 ⁇ .
  • Fig. 25 shows the vascular endothelial cells and feeder-free cells of the present invention shown in Example 7.
  • A Wright-Giemsa stained image (A) of blood cells prepared from serum-free cynomolgus embryonic stem cells under serum-free conditions (using K NOCKOUT (registered trademark) SR) Staining (shows myelin peroxidase staining (B), double staining of esterase (C). Various myeloid cells are observed. Scale bar indicates 20.
  • FIG. 26 shows vascular endothelial progenitor cells from human embryonic stem cells according to the “method for inducing differentiation into vascular endothelial cells and blood cells without a feeder” of the present invention shown in Example 8. And “specific progenitor cells” of blood cells (structures consisting of sac-like structures and spherical cell populations).
  • the scale bar indicates 100 ⁇ u m.
  • Fig.27 shows Wright-Giemsa staining image (A) of mature blood cells (floating cells) produced from the above-mentioned "specific progenitor cells” and special staining (esterase double staining (B), Neutrophil alkaline phosphatase staining (C)).
  • A mature blood cells
  • B specific progenitor cells
  • C Neutrophil alkaline phosphatase staining
  • Various myeloid cells are observed, ie cells at each stage of differentiation ranging from myeloblasts to mature blood cells (neutrophils and macrophages).
  • the scale lever shows 20.
  • Fig. 28 shows the results of the method of inducing differentiation into vascular endothelial cells and blood cells without feeders shown in Example 8 in Example 8 in the presence of fetal bovine serum from human embryonic stem cells.
  • the expression of CD34 and CD45 in the floating cells prepared in step 1 was confirmed by flow cytometry. It can be seen that almost all cells express the panblood cell marker CD45, and blood cell differentiation is induced with very high efficiency. Also, since about 10% of CD34 positive cells are detected, hematopoietic stem cells are also present!
  • FIG. 29 shows a lymphoprep (registration) from blood cells produced by the “method of inducing differentiation into vascular endothelial cells and blood cells without a feeder” of the present invention shown in Example 8.
  • This is the result of concentrating neutrophils by density gradient centrifugation using a trademark (made by Daiichi Chemicals Co., Ltd.).
  • B concentration
  • A force
  • neutrophils are stained blue-purple by neutrophil alkaline phosphatase staining.
  • FIG. 30 shows serum-free conditions (KNOCKOU) from human embryonic stem cells according to the “method for inducing differentiation into vascular endothelial cells and blood cells without feeder” shown in Example 9.
  • Flow cytometry analysis of CD45 expression in suspension cells made with T (registered trademark) SR It was confirmed by one. It can be seen that almost all cells express CD45, which is a pan-blood cell marker, and blood cell differentiation is induced with very high efficiency.
  • Figure 31 shows N-cad herin in three types of human primary cultured vascular endothelial cells (human umbilical vein endothelial cells (HUVE C), human microvascular endothelial cells (HMVEC), and human aortic endothelial cells (HAEC)).
  • the intracellular expression pattern of was examined by immunostaining. While vascular endothelial cells prepared from primate embryonic stem cells clearly show cell membrane localization (in Figure 14), N-cadherin cell membrane localization has already been lost in primary human vascular endothelial cells.
  • the power S is understood.
  • the scale bar shows 50 01.
  • FIG. 32 is a tumor tissue specimen (hematoxylin eosin staining) shown in Example 10.
  • Ectodermal component neuroepithelial cell; Fig. A, tooth enamel epithelium; Fig. D), mesoderm component (smooth muscle; Fig. B, dentin; Fig. D), endoderm component (gut epithelium; Fig.) b, secretory gland tissue; the three germ layers of Fig. c) are observed.
  • FIG. 33 is a tumor tissue specimen (hematoxylin eosin staining) shown in Example 11.
  • Ectodermal component neuroepithelial cell; Fig. A, pigment epithelium; Fig. B, sebaceous gland: Fig. H), mesoderm component (bone; Fig. D, adipocyte; Fig. E, cartilage; Fig. F), endoderm
  • Fig. C and Fig. G There are three germ layer components (secretory glands; Fig. C and Fig. G).
  • FIG. 34 shows VE-cadherin expression examined by immunostaining after fixing the sac structure shown in Example 12 with an acetone / methanol mixture.
  • the localization of VE-cadherin at the intercellular boundary is clear. Paving stone cells become distant and motility increases, and the membrane localization of VE-cadherin is unclear.
  • VE-cadherin is expressed intracellularly. These VE-cadherin-positive cells are! /, And the power of PCNA, which is a proliferation marker, is positive.
  • the large cells (arrows) of VE-cadherin-negative cells are negative for PCNA and do not proliferate! / I understand.
  • the scale bar indicates 100 ⁇ m.
  • FIG. 35 shows the expression of VE-cadherin by immunostaining in the structure shown in Example 12 in which the structure shown in FIG. 34 was peeled and collected as a lump and subcultured. is there .
  • the scale bar indicates 20.
  • FIG. 36 shows subcultures of the sac-like structure and paving stone cells shown in Example 12, and the expression of VE-cadherin and PECAM1 was measured with a flow cytometer over time. Is. The initial passage is;! ⁇ 20%, but as the number of passages progresses, the VE-cadherin / PECAMl positive rate increases.
  • FIG. 37A shows immunostaining of monkey embryonic stem cell-derived endothelial cells and human aortic smooth muscle cells shown in Example 12 with anti-smooth muscle actin (ACTA2) antibody or control IgG (FIG. 37A, upper panel).
  • the lower part of FIG. 37A shows a differential interference image of the cell.
  • the population of monkey embryonic stem cell-derived endothelial cells was ACTA2-negative, confirming that perisite was not contaminated.
  • the scale bar indicates 20.
  • FIG. 37B shows immunostaining of monkey embryonic stem cell-derived endothelial cells and human aortic smooth muscle cells shown in Example 12 with anti-platelet-derived growth factor receptor ⁇ (PDGFR ⁇ antibody or control IgG).
  • Figure 37B, upper panel shows a differential interference image of the cells, and the population of monkey embryonic stem cell-derived endothelial cells is PDGFR ⁇ -negative, and it is confirmed that the perisite is not contaminated.
  • the scale bar shows 20 ⁇ m.
  • FIG. 38A shows immunostaining of monkey embryonic stem cell-derived endothelial cells and undifferentiated embryonic stem cells shown in Example 12 with anti-human Nanog antibody (Reprocell) or control IgG (FIG. 38A, upper panel). ).
  • the lower part of FIG. 38A shows a differential interference image of the cell.
  • the monkey embryonic stem cell-derived endothelial cell population was Nanog-negative, and it was confirmed that undifferentiated embryonic stem cells were not contaminated.
  • the scale bar indicates 20.
  • FIG. 38B shows the anti-human Nanog antibody (left) using the lysate of human umbilical vein endothelial cells (HUVEC), monkey embryonic stem-derived endothelial cells and undifferentiated embryonic stem cells shown in Example 12. Western blotting was performed with anti- ⁇ -tubulin antibody (right). The population of monkey embryonic stem-derived endothelial cells was Nanog-negative, confirming that the undifferentiated embryonic stem cells were not contaminated.
  • HUVEC human umbilical vein endothelial cells
  • monkey embryonic stem-derived endothelial cells undifferentiated embryonic stem cells shown in Example 12.
  • Western blotting was performed with anti- ⁇ -tubulin antibody (right).
  • the population of monkey embryonic stem-derived endothelial cells was Nanog-negative, confirming that the undifferentiated embryonic stem cells were not contaminated.
  • FIG. 39A shows the results of FACSAria (BD Bioscience) after the passage of the sac-like structure shown in Example 12, after the JrL ⁇ E-cadherinirL body (Beckman Coulter, Clone Using TEA1.31), the VE-cadherin positive and negative fractions are sorted.
  • VE_ca Even when the dherin-positive fraction (Fig. 39A) was subcultured, VE-cadherin expression was stably maintained (Fig. 39B), and it was confirmed that the cells were amplified about 160 times by subculture for 5 times. It was.
  • FIG. 39C shows the expression of VE-cadherin by immunostaining using an anti-VE-cadherin antibody (BD, clone 75) after subculture of the VE-cadherin positive fraction shown in Example 12. It was confirmed. The localization of VE-cadherin was confirmed at the intercellular junction of all cells.
  • the scale bar shows 10 01.
  • FIG. 40 shows the results of sorting the sac-like structures shown in Example 12 into VE-cadherin positive and negative fractions using FACSAria (BD Bioscience) after one passage. It is.
  • the VE-cadherin negative fraction does not express VE-cadherin on the cell surface (Fig. A)
  • VE-cadherin is expressed in the cell, coding ability (Fig. B), and low acetylation.
  • Specific gravity lipoprotein (Ac-LDL) uptake capacity Di (Ac-LDL; fluorescently labeled Ac_LDL.
  • LDL is unlabeled LDL as a negative control
  • Figure C committed to vascular endothelial cells It was confirmed to be a cell.
  • the scale bar indicates 100.
  • FIG. 41a is a tumor tissue specimen of rat Dariooma transplanted into mice shown in Example 13.
  • the tumor is rich in large blood vessels and easily bleeding.
  • FIG. 41b shows the histological examination after fixing the tumor tissue of FIG. 41a with formalin, preparing a sliced slice, and performing hematoxylin-eosin staining (HE staining). . It can be seen that co-transplanted cynomolgus embryonic stem cell-derived vascular endothelial cells show a structure rich in blood vessels.
  • the scale bar in Figure 41b shows 100 mm.
  • Fig. 41c shows immunostaining of tumor cells formed by co-transplantation of rat Dario cell and cynomolgus monkey embryonic stem cell-derived vascular endothelial cells using anti-human HLA-A, B, and C antibodies. This is the result.
  • Endothelial cells lining the intratumoral neovascularization are derived from primates, that is, derived from vascular endothelial cells derived from power bicyno embryonic stem cells.
  • FIG. 42a shows the results of analysis of the expression of VE-cadherin and PECAM1 on the cell surface of the vascular endothelial cells induced in human embryonic stem cells shown in Example 14 using a flow cytometer.
  • the proportion of VE-cadherin / PECAMl double positive cells is about 20% in the early passage, but it reaches 70% in the later passage.
  • FIG. 42b is an in vitro functional evaluation of human embryonic stem cell-derived vascular endothelial cells shown in Example 14. It can be seen that both acetylated low density lipoprotein uptake and code formation are positive.
  • the scale bar indicates 100 m.
  • FIG. 42c shows the results of plug assembly for evaluating the function of human embryonic stem cell-derived vascular endothelial cells shown in Example 14 in vivo.
  • Collagen plugs transplanted with human embryonic stem cell-derived vascular endothelial cells were collected, fixed in formalin, and then immunostained with human HLA-A, B, C antibodies and human PECAM1 antibodies. It can be seen that the new blood vessels formed in the plug are derived from primates, ie, human embryonic stem cell-derived vascular endothelial cells.
  • the scale bar indicates 40 ⁇ m.
  • FIG. 43 shows the analysis of the expression of blood cell markers centered on neutrophils shown in Example 15 by flow cytometry.
  • FIG. 44 shows the results of measuring the positive rate of human-derived neutrophils by flow cytometry using anti-human CD66b antibody shown in Example 16.
  • the present invention is basically intended for the inventors to reduce the stress on cells and achieve the inherent controlled differentiation of cells during culture and passage of primate embryonic stem cells. It was achieved as a result of finding appropriate conditions. Specifically, as described later, the present inventors have found conditions for reducing or eliminating stress on embryonic stem cells, differentiated progenitor cells and mature cells of primates, and thereby the embryos. Achieving sustained and stable maintenance of sex stem cells, and at the same time, safely producing target cells that are efficient from embryonic stem cells The method to complete was completed.
  • the method of the present invention for culturing and passaging primate embryonic stem cells in an undifferentiated state comprises the following steps.
  • step (B) exfoliating the embryonic stem cell colonies formed in step (A) in the presence of a cell exfoliating agent
  • the embryonic stem cell colony obtained in the above step (B) is seeded in a medium containing a protein component in a container coated with an extracellular matrix under a feeder-free and site-free force-in.
  • the present inventors have experimentally confirmed that primate embryonic stem cells are in an undifferentiated state without supplementation with external specific factors (factors secreted by feeder cells, synthesis site force-in, etc.). It is based on having acquired the knowledge that it has the ability to maintain. Based on these findings, primate embryonic cells, which were difficult or impossible in the past, can be transformed into feeder-free, site-free by selecting an appropriate medium, culturing and subculturing with appropriate procedures. It was possible to maintain the undifferentiated state under force-in, and to continue to pass stably over a long period of several tens of times without causing chromosomal abnormalities.
  • the "primate embryonic stem cell” means an embryonic stem cell derived from any primate animal. Primate embryonic stem cells and methods for their preparation are known, for example, force cynomolgus monkey embryonic stem cells [Suemori, S., et al., “Strengthened cynomolgus blastocysts produced by IVF or ICSI. TeTA bstablishment or embryonic stem cell lines from cynomolgus monkey blastocysts produced by IVF or ICSL), Dev. Dynamic s Vol. 222, Brother pp. 273 p.
  • primary embryonic stem cell in the context of the present invention means an undifferentiated primate embryonic stem cell.
  • Confirmation that the primate embryonic stem cells are in an undifferentiated state can be performed by a known evaluation method. For example, expression of molecular markers (expression measurement by flow cytometry such as SSEA-4, Oct-4, immunostaining of Oct_4, Nanog, etc.), confirmation of pluripotent differentiation in in vitro experiments, immunodeficient mice, etc. Confirmation of the formation of teratomas by transplantation into the cerebral organ is performed by methods known to those skilled in the art.
  • the primate embryonic stem cell culture and passaging method uses a normal medium (with no addition of cytodynamic force) used to maintain primate embryonic stem cells. You can power s . Specifically, for example, Iskov modified Dulbecco medium (IMDM / Ham's F-12) and the like can be mentioned.
  • IMDM Iskov modified Dulbecco medium
  • the seeding of embryonic stem cells in the medium is performed according to the method described below. It should be noted that the medium used for the passage may not be necessarily the same as long as the embryonic stem cells can be maintained in an undifferentiated state. Specifically, the culture media in step (A) and step (C) may be the same or different.
  • the protein component used in the above-mentioned "method for maintaining undifferentiated primate embryonic stem cells” may be "other than animal serum” used to maintain primate embryonic stem cells.
  • animal serum used to maintain primate embryonic stem cells.
  • serum albumin for example, serum albumin, human type AB serum and the like can be mentioned.
  • commercially available serum-free additives suitable for maintenance and proliferation of embryonic stem cells such as KNOCK OUT (registered trademark) SR (manufactured by Invitrogen), can also be used.
  • the "extracellular matrix” includes the extracellular matrix components secreted by cells (Matrigel (registered trademark) Matrix (manufactured by BD), etc.) and other extracellularly secreted cells. Components that enhance, collagen, laminin, fibronectin, vitronectin, hyaluronic acid obtained from living organisms including humans, and artificially synthesized products of these proteins and polysaccharides (including degradation products and fragmented products), from living organisms including humans Examples thereof include serum and plasma obtained and products separated or purified from these.
  • the culture vessel can be coated with the extracellular matrix by a conventional method.
  • the extracellular matrix is preferably derived from primates including humans. In addition, it is preferable to use the latter when the extracellular matrix is obtained from both a heterogeneous animal and a human, and the latter is inferior to the former.
  • a method described below which is preferably performed by a method that does not induce cell stress to the maximum extent.
  • a culture dish coated with a Matrigel (registered trademark) matrix is used, desvases and the like are preferable, but it is not limited to this as long as there is no cell stress or it has been confirmed that it is sufficiently low.
  • the stress applied to the cells is easily determined based on the number of dead cells and the rate of cell proliferation. It is necessary to confirm that a chromosomal abnormality has been induced, etc.
  • the culture vessel may be any vessel that is usually used for cell culture.
  • the cell density when seeding primate embryonic stem cells in culture vessels is chosen to minimize stress on the cells. Control of cell density is achieved by appropriate selection of colony size and number of primate embryonic stem cells.
  • the size of a colony when seeded in a culture vessel of primate embryonic stem cells is easy to confirm by observation using a microscope (such as an inverted phase contrast microscope) and is highly effective. It may be confirmed by a method (visual observation, measurement of the amount of side scattered light, measurement of solution turbidity, etc.).
  • the optimal colony size is determined for each individual primate embryonic stem cell.For example, in the case of cynomolgus monkey embryonic stem cells or human embryonic stem cells, The diameter is about 100 m to about 2000 m, preferably about 300 m to about 1000 m, more preferably about 500 ⁇ m.
  • Control of the size of colonies when primate embryonic stem cells are seeded in a culture vessel is achieved by appropriately performing a cell detachment operation.
  • Cell detachment is preferably performed under conditions that minimize stress on the cells.
  • the release agent at least one selected from the group consisting of trypsin, collagenase and dispase is preferred, and dispase alone or a combination of dispase and other release agent is preferable.
  • a commercially available cell detachment agent (cell detachment solution for primate embryonic stem cells (Librocell)) or the like can also be used.
  • a suitable exfoliation method is a force determined for each primate embryonic stem cell.
  • the method shown in Example 1 described below is recommended.
  • a feeder-free culture of cynomolgus monkey embryonic stem cells In cell detachment at the time of passage, only extracellular matrix is used rather than collagenase solution (which may degrade cell membrane proteins).
  • collagenase solution which may degrade cell membrane proteins.
  • cell stress can be greatly suppressed, which is preferable, but not necessarily limited thereto.
  • the number of colonies when seeding in a primate embryonic stem cell culture vessel can be easily confirmed by observation with a microscope (such as an inverted phase contrast microscope). You can also use a calculator)!
  • the optimal value for the number of colonies (density) when seeded in a primate embryonic stem cell culture vessel is determined for each individual primate embryonic stem cell, but the colonies do not interact during the culture period. It is necessary to make it less than the density that does not fuse. For example, in the case of force cynomolgus embryonic stem cells or human embryonic stem cells, the methods shown in the examples described below (particularly Examples;! To 3) are recommended.
  • Passage frequency (timing) of primate embryonic stem cells is determined for each primate embryonic stem cell.
  • the colony diameter reaches approximately twice that of the seeding time. Is easy.
  • the methods shown in the examples described later are recommended. That is, the colony size is about 200 ⁇ m to about 4000 ⁇ m, preferably about 600 ⁇ m to about 2000 ⁇ m, more preferably about 1000 ⁇ m. When it reaches, it is peeled off and sown again.
  • exfoliation operation according to the method shown in Examples 1 to 3 is performed, colonies are dispersed into uniform colonies having an average diameter of about 500 m.
  • the primate embryonic stem cells are maintained in an undifferentiated state by exchanging the medium based on the above-mentioned colony size.
  • the specific frequency varies depending on the origin of embryonic stem cells, culture conditions, etc., and is adjusted as appropriate.
  • a good undifferentiated state is maintained by exchanging the medium 4 times or more, preferably 5 times or more, more preferably 6 times or more per week (7 days). Is done.
  • Culture conditions for primate embryonic stem cells may be any conditions suitable for culturing embryonic stem cells, for example, 37 ° C, 5% by volume, etc. The concentration may be changed.
  • Method for culturing and passage of primate animal cells” of the present invention can be cryopreserved by a conventional method.
  • a conventional method for example, in the case of cynomolgus monkey embryonic stem cells or human embryonic stem cells, the method shown in Example 1 described below is recommended.
  • the culturing operation in the method of the present invention is extremely simple and does not require special equipment or special skills, so it can be performed immediately at any facility in the world.
  • the primate embryonic stem cell undifferentiated maintenance culture method according to the present invention can be widely implemented in the three fields of “clinical medicine”, “medical industry”, and “basic medical biology research”. It can contribute greatly to these developments.
  • the present invention relates to a method for producing blood cells, myeloid cells, vascular endothelial progenitor cells, stromal cells, hematopoietic stem cells and the like from primate embryonic stem cells without a feeder.
  • the method for producing blood cells and / or vascular endothelial progenitor cells using the feeder-free device of the present invention comprises the following steps.
  • step (B) Step for producing specific progenitor cells including floating cells and adherent cells by adhesion culture of the embryoid body or embryoid body-like cell aggregate obtained in step (A) in the presence of site force-in.
  • step (C) Step of separating floating cells and adherent cells from the specific progenitor cells obtained in step (B).
  • the method for producing blood cells, hematopoietic stromal cells and / or hematopoietic stem cells with the feeder-free device of the present invention comprises the following steps.
  • step (B) a step of producing specific precursor cells including floating cells and adherent cells by adhesion culture of the embryoid body or embryoid body-like cell aggregate obtained in step (A) in the presence of site force-in, and
  • step (C) A step of culturing the specific progenitor cells obtained in step (B) while separating floating cells.
  • "Suspension culture” in the above-described method of the present invention means culturing cells while maintaining a floating state using a low-adsorption culturing vessel or the like (low-attachment plat es et.).
  • the “adhesion culture” in the above-mentioned method of the present invention is a culture using a normal cell culture vessel in a state in which adhesion of cells to the culture vessel is ensured.
  • the above-described methods (1) and (2) of the present invention basically consisted of the present inventors' "formation of embryoid stem cells or similar cell aggregates from embryonic stem cells. Under the conditions, it is based on the finding that, when adhesion culture is performed with an appropriate technique in an appropriate differentiation medium containing cytoforce-in, differentiation control into cells of the target series can be achieved. It has become possible to produce and expand reproduction of blood cells, vascular endothelial progenitor cells, stromal cells, hematopoietic stem cells, myeloid cells, etc. from primate embryonic stem cells that were difficult or impossible in the past.
  • embryonic stem cells differentiate into vascular endothelial cells and blood cells via undifferentiated mesoderm, common precursor cells of vascular endothelial cells / blood cells (for example, Jun Yamashita, Inflammatory Disease) .Reproduction 22, Vol. 509, 2002).
  • blood cells refer to all blood cells.
  • specific examples of the blood cells include, for example, hematopoietic stem cells 0102, lymphoid stem cells 0133, and lymphoid dendritic cells in the differentiation system from embryonic stem cells to blood cells shown in FIG.
  • the “blood cell” includes a progenitor cell of a hematopoietic stem cell; It includes all forms of blood cells that exist on all differentiation processes up to differentiation into treetop blood.
  • myeloid cells examples include myeloblasts, promyelospheres, myelospheres, retromyelocytes, neutrophils, monocytes, macrophages and the like.
  • the origin of the primate embryonic stem cells that are the starting material is not particularly limited, and is maintained by passage according to the method described in I above and, if necessary, cryopreserved! I'll use it with power.
  • the culture technique in the above-mentioned "11. Method for inducing differentiation of primate embryonic stem cells without feeder” is the formation of embryoid body or embryoid body-like cell aggregate by suspension culture of primate embryonic stem cells. And a step related to the adhesion culture of a cell aggregate of embryoid body or embryoid body-like body.
  • the “cell aggregate similar to embryoid body” means a cell aggregate in the middle of formation of an embryoid body from embryonic stem cells.
  • Examples of methods for forming a cell aggregate of embryoid bodies or embryoid body-like cells include a conventional hanging drop method, a culture using a conventional non-adhesive culture dish, and a culture using a conventional semi-solid medium.
  • the present invention is not limited thereto as long as a definitive body or a cell aggregate similar to a definitive body is formed.
  • the period of suspension culture for producing embryoid body or embryoid body-like cell aggregates varies depending on the cells, culture conditions, and desired product. Usually, it is about 2 days to 2 weeks. The shorter the period, the greater the ratio of cell clumps to embryoid bodies.
  • the pulmonary lobe or similar cell aggregates are adhered and cultured in an optimized differentiation medium as they are. This is different from the conventional method of refining and separating the “progenitor cell population” that has been differentiated in the preferred direction after loosening to the cellular level from the embryoid body or similar cell aggregates by enzyme treatment etc. It is epoch-making in that differentiation induction can be achieved simply and safely with high efficiency.
  • the target series can be obtained with extremely high efficiency (efficiency of almost 100% for both vascular endothelial cells and blood cells) Differentiation control is achieved. This shows that the maintenance of “controlled differentiation” in the development process of individual animals is achieved.
  • “Differentiation medium” means a medium in which at least one cytodynamic in is added to a medium for culturing the above-described primate embryonic stem cells in an undifferentiated state.
  • the medium may contain other suitable additives as desired, as long as they do not adversely affect cell maintenance and differentiation.
  • the "site force in” can be appropriately selected according to the purpose, and such a cytokin is known to those skilled in the art.
  • the cyto force-in that can be used in the present invention is not particularly limited as long as it is a factor for differentiating embryonic stem cells into blood cells and / or vascular endothelial cells.
  • stem cell factor SCF
  • G—CSF granulocyte colony Stimulating factor
  • GM—CSF granulocyte macrophage colony stimulating factor
  • M—CSF macrophage colony stimulating factor
  • EPO erythropoietin
  • TPO thrombopoietin
  • FL Flt3 ligand
  • IL interleukin
  • VEGF vascular endothelial growth factor
  • BMP bone morphogenetic protein
  • BMP eg BMP-4, etc.
  • oncostatin M acidic and basic fibroblast growth factor (acidic) FGF, basic FGF), angioboyetin family (for example, Angiopoietin-1 and Angiopoietin-2), and the like.
  • the G-CSF has a function of enhancing neutrophil production.
  • EPO erythropoietin
  • TPO thrombopoietin
  • NK cells natural killer cells
  • the basic culture component used in the above-mentioned "11. Method for inducing differentiation of primate embryonic stem cells without feeder” according to the present invention is as described above from primate embryonic stem cells to vascular endothelial cells.
  • Specific examples of the medium that can be used for inducing differentiation into blood cells include Iskov modified Dulbecco medium (MDM).
  • Method for inducing differentiation of primate embryonic stem cells without feeder includes primate embryonic stem cells to vascular endothelial cells 'blood cells'
  • fetal calf serum, human serum (reducing the risk of inducing immune rejection, type AB serum should be used if it is suitable for inducing differentiation into Are preferable)
  • KNOCKOUT (registered trademark) SR manufactured by Invitrogen, and the like.
  • the coating component of the culture dish used in the above-mentioned "11. Method for inducing differentiation of primate embryonic stem cells without feeder” includes primate embryonic stem cells to vascular endothelial cells and blood cells.
  • a specific example is gelatin that is suitable for inducing differentiation.
  • the culture vessel may be any vessel that is usually used for cell culture.
  • the primate embryonic stem cell culture conditions may be any conditions suitable for culturing embryonic stem cells, for example, 37 ° C, 5% by volume, etc. You may be fi.
  • the culture conditions in the “method for inducing differentiation of primate embryonic stem cells without feeder” according to the present invention are appropriately set according to the type of primate embryonic stem cells used.
  • Force can S, for example, 37 ° C, 5% by volume Ji ⁇ second conditions.
  • adhesion culture is performed until “specific progenitor cells” are formed.
  • the medium exchange and detachment operation are performed according to the description in “1. Method for inducing differentiation of primate embryonic stem cells without feeder”.
  • Specific progenitor cells refer to embryoid bodies or cells that contain floating cells (spherical or near-spherical cells that have the property of floating in the culture medium) and adherent cells (cells that adhere to the culture vessel). Means a progenitor cell differentiated from an agglomerate-like cell aggregate. This may form a sac-like structure containing spherical cell populations and adherent cells as floating cells (containing the spherical cell population inside), but sac-like structures are not necessarily formed. As described in the examples below, by culturing embryoid bodies or cell aggregates similar to embryoid bodies under the appropriate conditions in the presence or absence of serum, the sac structure is included or not included. Specific precursor cells can be formed.
  • spherical cell When a sac-like structure is formed, floating cells (spherical cells) are released not only in the structure but also in the culture solution. If the sac-like structure is not formed, it exists in a suspended state in the culture medium. Therefore, in this specification, the phrase “spherical cell” or “floating cell” broadly refers to both forms of cells. Blood cells (such as hematopoietic progenitors and mature blood cells committed to myeloid cells) can be derived from floating cells (spherical cells) of specific progenitor cells, and vascular endothelial progenitor cells and hematopoietic cells can be derived mainly from adherent cells. Stromal cells can be induced. Hematopoietic stem cells are derived from both floating cells and adherent cells.
  • the selection of “progenitor cell population” that is being differentiated into a specific lineage is preferably performed based on observation of the tissue morphology of cells under a phase contrast microscope. Achieving “correctly controlled and effective differentiation induction” by a method based on the tissue morphology rather than the conventional method of sorting cells using a cell sorter such as a cell sorter using specific antibodies against molecular markers That power S. That is, in the case of the conventional method based on molecular markers, there is a problem that cells are damaged by the use of a cell separation device whose specificity is always high.
  • the various embryonic morphology is determined by observing under a phase-contrast microscope while continuing adhesion culture in a new culture dish without destroying the embryoid body. And sort.
  • the sac-like shape as shown in Fig. 12, Fig. 19 and Fig. 26
  • the “structure” and the spherical cells (floating cells) contained therein are the precursors of vascular endothelial cells and blood cells.
  • the method for producing blood cells and / or vascular endothelial progenitor cells without feeders according to (1) Floating cells and adherent cells are separated from specific progenitor cells including vascular and adherent cells, and blood cells and vascular endothelial progenitor cells are prepared from each.
  • the floating cells in the culture solution and the spherical cells in the sac structure are separated by centrifugation or the like. Separation of the spherical cells from the sac-like structure is carried out by providing an opening in the sac-like structure and suspending the spherical cells by an appropriate method. This operation is preferably performed before the sac-like structure is completely filled with cells. Normally, the opening of the sac-like structure is closed again by culture, and the inside is filled with spherical cells.
  • the method for producing blood cells, myeloid cells, hematopoietic stromal cells and / or hematopoietic stem cells without feeders in (2) can be performed by appropriately separating floating cells released into the culture medium. And culturing specific progenitor cells having a sac-like structure containing floating cells and adherent cells as they are.
  • the suspension cells are separated by centrifugation or the like.
  • blood cells are continuously cultured in a mixed state of adherent cell populations containing sac-like structures and floating cell populations while appropriately releasing floating cells without separating spherical cells from sac-like structures.
  • hematopoietic stem cells can be obtained. Formation of the generated stromal cells or hematopoietic stem cells can be confirmed by detection of each cell marker. Such markers are generally known to those skilled in the art.
  • an adherent cell (blood cell progenitor) is obtained by a method in which a blood cell progenitor cell (such as a spherical cell in a sac-like structure) produced by adhesion culture of embryoid body cells is further cultured and subcultured.
  • a culture method for expanding and reproducing blood cells is also included, while maintaining the situation where cells, hematopoietic stroma cells) and floating cells (blood cells) are mixed.
  • the present invention provides for the first time a method for producing “hematopoietic stromal cells” from embryonic stem cells. It has also been clarified that the hematopoietic stem cells produced according to the present invention have not only properties as floating cells but also properties as adherent cells (see FIG. 23).
  • the vascular endothelial cells, blood cells, myeloid cells, hematopoietic stromal cells, hematopoietic stem cells, etc. of the present invention are, for example, a medium such as a cell cryopreservation solution such as bun bunker (manufactured by Nippon Genetics), and nitrogen. Maintained under gas freezing conditions.
  • a cell cryopreservation solution such as bun bunker (manufactured by Nippon Genetics)
  • bun bunker manufactured by Nippon Genetics
  • Vascular endothelial cells, blood cells, myeloid cells, hematopoietic stromal cells, hematopoietic stem cells and the like obtained by such a production method are substantially free from contamination with heterologous animal cells, infection with heterologous animal-derived viruses, and the like. Excellent properties are shown.
  • the vascular endothelial cells, blood cells, and hematopoietic stroma cells according to the present invention all exhibit high purity and homogeneous properties. Accordingly, the vascular endothelial cells according to the present invention relate to materials for the treatment of vascular injury and improvement of local blood flow, transplant materials, the use for the production of these materials, and the generation and differentiation mechanism of vascular endothelial cells.
  • the blood cells according to the present invention can be used as blood for transfusion, use for production of blood for transfusion, and materials for basic research on hematopoietic mechanism.
  • the hematopoietic stromal cells according to the present invention can be used as materials for transplantation medicine for hematopoietic disorder and basic research on hematopoietic mechanisms.
  • myeloid cells are considered useful for the treatment of bone marrow injury.
  • vascular endothelial precursors From the cells, a cell population with a higher maturity can be separated as a VE-cadherin-positive PECAM1-positive double-positive population on the cell membrane surface by cell sorter or bead sedimentation. Specifically, vascular endothelial cells are separated by, for example, cell sorting by flow cytometry using a specific antibody against a marker such as VE-cadherin or PECAM1, cell sorting using a magnetic bead holding the antibody, etc. Can do.
  • vascular structure it is also possible to obtain a three-dimensional vascular structure by culturing in, for example, a collagen gel using the vascular endothelial cell of the present invention. It is also possible to form a new vascular network in vivo by transplanting the vascular structure thus constructed to an animal.
  • a flow using an antibody against a lineage-specific marker using blood cells produced by the method for producing blood cells without a feeder of the present invention.
  • Cell sorting by cytometry, cell sorting using magnetic beads holding the antibody, etc. can be applied to separate and concentrate only a specific series of blood cells such as hematopoietic stem cells, neutrophils, monocytes, lymphocytes, etc. it can.
  • hematopoietic stem cells are separated and concentrated by collecting a CD34 positive CD45 positive double positive cell fraction using CD34 antibody and CD45 antibody.
  • a feeder-free differentiation medium in which hematopoietic stem / progenitor cells may be further differentiated under appropriate conditions according to the type of the target blood cell or the like. You may change the inside site power in appropriately.
  • examples of blood cell production by various site force-in include differentiation into granulocytes by G-CSF and GM-CSF, and simple production by GM-CSF and M-CSF.
  • Sphere / Macropha Differentiation into NK cells by IL 15 differentiation into red blood cells by EPO, differentiation into megakaryocytes / platelets by TPO, differentiation into dendritic cells by IL 4 and GM-CSF, etc. It is done.
  • the method for maintaining undifferentiated primate embryonic stem cells under feeder-free and site-free force-in, and differentiation into vascular endothelial cells, blood cells, etc. without feeders according to the present invention.
  • the induction method is not limited to use with embryonic stem cells, and various cells (such as testicular stem cells, adult stem cells, etc. that retain pluripotent differentiation potential) can be modified by modifying the culture conditions. Application development is possible as a differentiation technique to group).
  • a primate animal can be obtained by a simple culturing operation and using only an inexpensive culturing device without being substantially contaminated with a heterologous animal cell or infected with a heterologous animal-derived virus.
  • embryonic stem cells From embryonic stem cells, vascular endothelial cells that can be maintained for passage, blood cells that can be expanded and regenerated (including hematopoietic stem cells to mature blood cells), hematopoietic stroma cells, etc. are all very high and efficient (almost) It is possible to create it near 100%!
  • the “method for inducing differentiation into vascular endothelial cells and blood cells without a feeder” according to the present invention has very high differentiation efficiency, no cell damage, and high feasibility. Implemented promptly around the world, the benefits are enormous.
  • Forced Cynomolgus Embryonic Stem Cell is an undifferentiated maintenance culture solution 1 1 (composition: DMEM / H ⁇ S F-12 (manufactured by Kojin Bio), 20 vol% KNOCKOUT (registered trademark) SR (Invitrogen Corp.) ImM L gnoretamine (Invitrogen Corp.), 2 mM non-essential amino acid solution (Invitrogen Corp.), ImM sodium pyruvate (Invitrogen Corp.), final concentration 100 U / ml Penicillin (Invitrogen Corp.), final concentration 100 g / ml 10 cm culture dish coated with leptomycin (manufactured by Invitrogen Corp.) using Matrigel® matrix (manufactured by BD (BD Biosciences)) diluted 30 times at room temperature for 15-30 minutes The cells were cultured on a top or on a 78 cm 2 culture dish in a CO incubator at 37 ° C and 5 volumes 0 / oCO.
  • Forced cynomolgus embryonic stem cells have a relatively uniform colony size of 500 m in diameter, and are in the field of view of a 4 ⁇ objective lens (10 ⁇ eyepiece) of a phase contrast microscope; ! ⁇ Seeding at a density of about 2 pieces.
  • the medium is changed on the next day. Further, since the size of the colony becomes about 1000 on the next day, it is detached with dispase ⁇ 0 (80 Biosciences)] and subcultured to a new culture vessel coated with Matrigel (registered trademark) matrix.
  • the force cynomolgus embryonic stem cells can be fed to the above-mentioned culture medium without adding feeders that do not allow colonies to join, and to which the synthetic site force-in is not added. It is possible to carry out amplification culture while maintaining an undifferentiated state appropriately.
  • the undifferentiated maintenance state was maintained appropriately. Specifically, the cell morphology preferred as an undifferentiated state (see FIG. 1) and the high level of undifferentiated maintenance markers SS EA-4, Oct-4, Nanog, Tra-1-60, Tra-1-81 Expression (see FIGS. 2 and 3) and tumor formation in immunodeficient mice (SCID mice) (see FIGS. 4 and 5) were confirmed.
  • FIG. 1 shows colonies of force cynomolgus embryonic stem cells cultured by the above method.
  • A shows cells that were passaged 20 and B was frozen at thirty-five passages and thawed one more time after thawing.
  • Fig. 2 shows the expression of SSEA-4 and Oct-4, which are undifferentiated maintenance markers, in flow cytometry in the 20th-generation force-drunken monkey embryonic stem cells cultured by the above method. It is the result measured by Lee. In both cases, very high! /, Expression (> 95%) is confirmed.
  • Figure 3 shows the expression of undifferentiated maintenance markers, Tra-1_60, Tra-l_81, and Nanog, by immunostaining in the 20th-passed force-ducked monkey embryonic stem cells cultured as described above. It is a thing. The expression of both markers is confirmed in almost all cells.
  • Figure 4 shows the testis photographed two months after transplanting the 21st-generation viable cynomolgus embryonic stem cells cultured by the above method under the testis capsule of three immunodeficient mice (SCID mice). It is. Teratoma formation was confirmed in all three animals.
  • Fig. 5 is a tissue specimen of the above tumor (hematoxylin eosin staining). Neural epithelium, teeth, secretory glands, gut-like epithelium, smooth muscle, etc. are observed as described.
  • Example 4 and Example 7 it is possible to produce vascular endothelial cells and expand and regenerate blood cells and hematopoietic stromal cells from the power-dwelling monkey embryonic stem cells maintained undifferentiated according to the present invention.
  • Thaw frozen cells in the following procedure add 1 ml of the medium for maintaining undifferentiation that has been pre-warmed to 37 ° C to a freezing cynomolgus embryonic stem cell freezing tube, rapidly thaw by pipetting, and then add 15 ml conical to the cell suspension. Transfer to a tube and collect cells by sedimentation (1000 rpm, 5 minutes, 4 ° C). After suspending the cells in an undifferentiated maintenance medium, The state of the cells is confirmed with a microscope and cultured in a culture vessel coated with Matrigel (registered trademark) matrix in a CO incubator at 37 ° C, 5 volumes 0 / oCO.
  • Matrigel registered trademark
  • Human embryonic stem cells consist of undifferentiated maintenance culture medium 12 (composition: DMEM / Ham's F_12 (manufactured by Kojin Bio), 20 vol% KNOCKOUT (registered trademark) SR (manufactured by Invitrogen Corp.), ImM L glutamine (Invitrogen) (Manufactured by Invitrogen Corp.), 2 mM non-essential amino acid solution (manufactured by Invitrogen Corp.), 0.1 M 2-mercaptoethanol (manufactured by Sigma Chemical Co.), final concentration lOUU / ml penicillin (invitrogen) (Manufactured by Invitrogen Corp.) and a final concentration of 100 g / ml streptomycin (manufactured by Invitrogen Corp.)) and Matrigel (registered trademark) Matritus (manufactured by BD Biosciences) diluted 30-fold.
  • the cells were cultured on a 10 cm culture dish or
  • Human embryonic stem cells have a relatively uniform colony size of 500 m in diameter, and have a field of view of a 4 ⁇ objective lens (10 ⁇ eyepiece) of a phase contrast microscope. Seed at a density of about three, and thereafter change the medium daily. After 3 to 4 days, the colony size force S reaches about 1000 m.
  • the stripping solution 1 composition: 0.25% trypsin solution (Invitrogen Corp.)
  • 1 mg / ml collagenase IV [Invitrogen (Invitrogen Corp.), 1% KNOCKOUT® SR (manufactured by Invitrogen Corp.), 1 mM calcium chloride (manufactured by Sigma Chemical Co.) based on phosphate buffer, or Peel off with primate embryonic stem cell detachment solution (Librocell) and pass to a new culture container coated with Matrigel® matrix
  • the specific procedure for peeling is as follows. Immerse the exfoliation solution in human embryonic stem cells from which the culture medium has been removed and react at 37 ° C for 5 minutes.
  • Fig. 6 shows a phase contrast micrograph of colonies of human embryonic stem cells of passage 24 cultured by the above method.
  • FIG. 7 shows the results of measurement of the expression of SSEA-4 and Oct-4, which are undifferentiated maintenance markers, in human embryonic stem cells of the 20th passage cultured by the above method by flow cytometry. In both cases, very high! /, Expression (> 95%) is confirmed.
  • FIG. 8 shows the expression of Oct-4 (A) and Nanog (B), which are undifferentiated maintenance markers, in human embryonic stem cells of the 25th passage cultured by the above method by immunostaining. It is confirmed that almost all cells have the ability to express both proteins!
  • FIG. 9 is a chromosome analysis diagram (G band method) of human embryonic stem cells.
  • the left is maintained by the conventional culture method recommended by the establishment organization (co-culture method using fetal mouse fountain fibroblasts as one feeder cell)! This is the result of the 20th passage in the “Culture method with one site-free force-in”. It was confirmed that no chromosomal abnormality occurred.
  • cryopreservation and thawing in liquid nitrogen can be performed by the method described in Example 1 using a commercially available cryopreservation solution for primate embryonic stem cells (Ribrocell).
  • a commercially available cryopreservation solution for primate embryonic stem cells Rosettacell
  • the human embryonic stem cells maintained undifferentiated in Example 2 proliferate while maintaining an undifferentiated maintenance state well after freezing and thawing.
  • Example 2 The medium described in Example 2 was used.
  • Human embryonic stem cells have a relatively uniform colony size of 500 m in diameter, and have a field of view of a 4 ⁇ objective lens (10 ⁇ eyepiece) of a phase contrast microscope. Seed at a density of about three, and thereafter change the medium daily. After 3 to 4 days, the colony size force S reaches about 1000 m.
  • the stripping solution 1 composition: 0.25% trypsin solution (Invitrogen Corp.)
  • 1 mg / ml collagenase IV [Invitrogen (Invitrogen Corp.), 1% KNOCKOUT® SR (manufactured by Invitrogen Corp.), 1 mM calcium chloride (manufactured by Sigma Chemical Co.) based on phosphate buffer, or
  • human AB serum which is exfoliated with primate embryonic stem cell detachment solution (Librocell) and coated with 5 gm 2 of fibronectin (BD) obtained from human plasma New culture container coated with laminin (Sigma) obtained from human placenta with 5 gm 2 and vitronectin (BD) obtained from human plasma 0.2 ⁇ g New coated with m 2
  • a new culture vessel, or a new culture vessel coated with 5 ag m 2 of type IV collagen (BD) obtained from human placenta is subcultured.
  • the specific procedure for peeling is as follows. Immerse the human embryonic stem cells after removing the culture medium and incubate the exfoliation solution at 37 ° C for 5 minutes. Then, aspirate the exfoliation solution and add DMEM / Ham's F-12. Allow to react for minutes. The cells are then detached and suspended by tapping the culture vessel and then suspended twice with a 1000 a 1 pipette tip. Collect the vesicles in a centrifuge tube, and sediment the cells by centrifugation (1000 rpm, 5 minutes, 4 ° C). Through the above operations, colonies of human embryonic stem cells are dispersed in a relatively uniform size of 500 m in diameter. By performing the above operation twice a week, human embryonic stem cells were appropriately maintained in an undifferentiated state in the culture medium described in Example 2 with no feeder and no addition of synthetic site force-in. Passage maintenance is possible.
  • the undifferentiated maintenance state is appropriately maintained even after the fourth passage.
  • the cell morphology preferred as an undifferentiated state and high expression of SSEA-4 and Oct-4 as undifferentiated maintenance markers were confirmed. See Figure 10.
  • FIG. 10 shows a culture dish (A) coated only with human-derived fibronectin (5 gm 2 ) and a culture dish (C) coated only with human AB serum by the above method.
  • 4 is a phase contrast micrograph of human embryonic stem cells at the 4th passage. V, the ability to hold the undifferentiated shape of the gap S
  • SSEA-4 and Oct-4 was measured by flow cytometry in these cells, high expression of both markers was confirmed in both cells (B, D).
  • IMDM Iscove's modified Dulbeccos medium
  • PAA Heat inactivated Fetal bovine serum
  • lmM / 3-Menolecaptoethanol Sigma Chemical Co.
  • 2 mM L-glutamine Invitrogen Corp.
  • VEGF vascular endothelial growth factor
  • BMP—4 20 ng / ml bone morphogenetic protein 4
  • SCF stem cell factor
  • 10 ng / ml Flt3 ligand The final concentration of 20 ng / ml interleukin 3 (IL 3) and the final concentration of 10 ng / ml interleukin 6 (IL6) ⁇ were added.
  • the culture was further filled with sterilized water, and suspended in a CO incubator at 37 ° C and 5 vol% CO for 3 days. After 3 days, the formation of cell clumps can be confirmed macroscopically, so that the lid surface of the culture dish was washed and collected, and coated with 0.1% gelatin (manufactured by Sigma Chemical Co.). Adhesion culture was performed on a culture dish (diameter 10 cm or 6 cm) at 37 ° C and 5% by volume in a CO incubator using differentiation medium 11 (added with site force in). Thereafter, the medium was changed every 3 to 4 days.
  • FIG. 12 shows vascular endothelial progenitor cells and blood cells created from cynomolgus monkey embryonic stem cells in the presence of fetal calf serum using a feeder-free vascular endothelial cell and blood cell differentiation induction and expanded reproduction technology.
  • Specific progenitor cells structures consisting of sac-like structures and globular cell populations).
  • the structure of the sac structure itself is not destroyed using a microknife (Stem eel 1 knife, manufactured by SweMed, etc.).
  • a microknife Stem eel 1 knife, manufactured by SweMed, etc.
  • the inner spherical cells were released slowly into the culture medium.
  • the viability of the spherical cells decreases.
  • the supernatant is collected by centrifuging and collecting the culture supernatant, while the sac-like structure and cells spreading from it are trypsin / EDTA solution (manufactured by Invitrogen Corp.)).
  • the strip was collected by reacting at 5 ° C for 5 minutes.
  • spherical cells were collected using a colony assembly kit (Methocult (registered trademark) GF + H4535 (Stemcell Technologies In) using a semi-solid medium containing methylcellulose. Blood cell production was confirmed by performing hematopoietic colony assembly (see FIG. 13).
  • Fig. 13 shows spherical cell force, Wright-Giemsa staining (A), special staining (myeloperoxidase staining (B) and esterase double staining (C)) of mature blood cells produced! .
  • Various myeloid cells are observed, ie cells in each stage of differentiation ranging from myeloblasts to mature blood cells (neutrophils and macrophages).
  • the collected sac-like structures and the cells spreading in the periphery are a new culture dish (diameter 10 cm or 6 cm) coated with 0.1% gelatin (manufactured by Sigma Chemical Co.). —1 (Site force-in added) was used for culture. Thereafter, the cells were detached every 3 to 4 days with trypsin / EDTA solution and subcultured 8 times with a dilution of about 1/3 (see FIGS. 14 to 16).
  • Figure 14 immunizes the expression of VE-cadherin, a vascular endothelial cell-specific marker, and N-cadherin, a vascular endothelial cell marker, of vascular endothelial cells produced from the above-mentioned "specific progenitor cells". It is the result examined by the dyeing test. As shown in FIG. 14, expression of VE-cadherin, a vascular endothelial cell-specific marker, was confirmed in almost all cells after 2 passages. Furthermore, as shown in this figure, N-cadherin, an adhesion factor known to be expressed in vascular endothelial cells, is also expressed in almost all cells by immunostaining and has a clear cell membrane localization. It was shown to admit. This is a very interesting finding as shown in Fig. 31 because the primary vascular endothelial cells obtained from the living body have been lost (see Fig. 14 and Fig. 31 for comparison).
  • Figure 31 shows N-cadherin in three types of commercially available human primary cultured vascular endothelial cells (human umbilical vein endothelial cells (HUV EC), human microvascular endothelial cells (HMVEC), and human aortic endothelial cells (HAEC)). It is the result of investigating the intracellular expression mode by immunostaining. From Fig. 31, it can be seen that the cell membrane localization of N-cadherin has already disappeared in commercially available human primary cultured vascular endothelial cells. On the other hand, vascular endothelial cells produced by the method of the present invention show clear cell membrane localization and correctly reflect the properties of vascular endothelial cells in the living body. (In Figure 14).
  • HMVEC human umbilical vein endothelial cells
  • HMVEC human microvascular endothelial cells
  • HAEC human aortic endothelial cells
  • Figure 15 shows the expression of PECAM1, which is a mature vascular endothelial cell marker, in vascular endothelial cells produced from the above-mentioned "specific progenitor cells". This was confirmed by flow cytometry by double staining with VE-cadherin, an experimental marker.
  • the horizontal axis represents PECAM1, and the vertical axis represents the expression intensity of VE-cadherin.
  • 40% of cells express both proteins, VE-cadherin, a pancreatic endothelial cell marker-specific marker for vascular endothelial cells, and PECAM1, a mature vascular endothelial cell marker, at the cell membrane level. It was confirmed that the above existed.
  • Figure 16 shows the code-forming ability (A) and the uptake ability of acetylated low-density lipoprotein (B) to confirm the maturation function of vascular endothelial cells produced from the above-mentioned “specific progenitor cells”. It is. In both cases, the acquisition of mature function is confirmed with high efficiency. Based on the above, the expression of VE-cadherin and PECAM1 is maintained even after 8 passages, and as shown in Fig. 16, vascular endothelium such as code-forming ability, acetylated low-density lipoprotein uptake ability, etc. Functional maturation of the cells was also confirmed.
  • IMDM Iscove's modified Dulbecco's medium
  • SR Invitrogen Corp.
  • ImM / 3-mercaptoethanol Sigma Chemical Co.
  • 2 mM L-Glutamine Invitrogen Corp. ⁇ 20 ng / ml vascular endothelial growth factor (VEGF), final concentration 20 ng / ml bone morphogenetic protein 4 (BMP—4), 20 ng stem cell factor (SCF), final concentration 10 ng / ml Flt3—ligand, final concentration 20 ng / ml interleukin 3
  • IL3 medium supplemented with
  • a cell aggregate cluster similar to the embryoid body is prepared by a hanging 'drop method using differentiation medium 1 2 (added with site force in). Specifically, after collecting force cynomolgus embryonic stem cells with a stripping solution, further react with 0.25% trypsin solution (manufactured by Invitrogen Corp.) at 37 ° C for 5 minutes to obtain a single cell level. To disperse. Suspend 3000 force cynomolgus monkey embryonic stem cells in 30 1 differentiation medium 1-2 (with site force in) and use a micropipette to place the back of the lid of a 10 cm diameter culture dish. Spot (2030 spots can be spotted in one culture dish).
  • the culture was further filled with sterilized water, and suspended in a CO incubator at 37 ° C and 5 vol% CO for 3 days. After 3 days, the formation of cell agglomerates can be confirmed macroscopically, so that the lid surface of the culture dish was washed and collected, and coated with 0.1% gelatin (manufactured by Sigma Chemical Co.). Adhesion culture was performed on a culture dish (diameter 10 cm or 6 cm) in a CO incubator using differentiation medium 12 (added with cyto force in) at 37 ° C and 5% by volume. Thereafter, the medium was changed every 3 to 4 days. Agglomerates of cynomolgus monkey embryonic stem cells continued to grow while spreading on a flat surface.
  • FIG. 17 shows mature blood cells created from serum-free cynomolgus embryonic stem cells in serum-free culture conditions (using KNOCKOUT® SR).
  • A Wright-Giemsa staining
  • B myelin peroxidase staining
  • C esterase double staining
  • FIG. 18 shows the results of the flow cytometry test at the third passage. It has been confirmed that there are several% or more cells in which VE-cadherin and PECAM1 are expressed in the cell membrane, which was previously considered to be unable to induce differentiation of endothelial cells without serum (see Fig. 18).
  • Differentiation medium 1 3 Composition: Knockout D-MEM (Invitrogen Corp., 20 wt% heat-inactivated fetal bovine serum (PAA Laboratories GmbH)], 0.1 mM / 3—Menolecaptoethanol (Sigma Chemical Co.), 1% non-essential amino acid solution (Invitrogen Corp.), ImM L gnoretamine (Invitrogen Corp.) Manufactured)] at a final concentration of 50 ng / ml bone morphogenetic protein 4 (BMP-4), 300 ng / ml stem cell factor (SCF), final concentration of 300 ng / ml Flt3 ligand, final concentration of 10 ng / ml interleukin 3 (IL3), A final concentration of 10 ng / ml interleukin 6 (IL6) and a concentration of 50 ⁇ g / ml granulocyte colony-stimulating factor (G-CSF)) were added.
  • BMP-4 bone morph
  • Example 1 Forced cynomolgus embryonic stem cells maintained undifferentiated in Example 1 were treated with collagenase IV (room temperature, 20 minutes) followed by chelating agent treatment (non-enzymatic cell dissociati on buffer [Invitrogen Corp. manufactured by Invitrogen Corp.) ] At room temperature for 20 minutes), and the force of the culture vessel coated with Matrigel (registered trademark) was peeled off. By this operation, the force cynomolgus embryonic stem cells were loosened in a state close to the individual cell level.
  • collagenase IV room temperature, 20 minutes
  • chelating agent treatment non-enzymatic cell dissociati on buffer [Invitrogen Corp. manufactured by Invitrogen Corp.)
  • Matrigel registered trademark
  • differentiation medium 1-3 non-adhesive culture vessels (6 cm diameter culture dish, etc.) coated with poly (2-hydoxyethyl metha crylate) (Sigma), or Hydrocell (CellSeed was used for suspension culture all day and night.
  • the medium is changed to the differentiation medium 1-3 added with the above-mentioned cyto force in (without the addition of cyto force in), and floating culture is further performed for about 2 weeks, so that the embryoid body (or a cell aggregate similar to embryoid body) is obtained.
  • the embryoid body or a cell aggregate similar to embryoid body
  • the embryoid body or cell aggregates similar to the embryoid body) collect the culture supernatant, centrifuge the cell components, and newly prepare the differentiation medium 1
  • the suspension was suspended in —3 (non-supplemented calorie) and transferred to a newly prepared non-adhesive culture vessel to continue suspension culture.
  • the embryoid body formed above is collected from the culture supernatant by centrifugation and differentiated on a culture dish (24-well multi-well dish) coated with 0.1% gelatin (manufactured by Sigma Chemical Co.). Using culture medium 13 (added with cyto force in), adhesion culture was performed at 37 ° C and 5% by volume in a CO incubator. Thereafter, the medium was changed every 3 to 4 days. Aggregates of force cynomolgus embryonic stem cells continue to grow while spreading on a flat surface, and after about 2 weeks, as shown in Fig. 19B, "specific precursor cells" (A structure consisting of a sac-like structure and a spherical cell population contained therein) was formed (one was formed for each aggregate). Fig.
  • FIG. 19 shows a phase contrast micrograph of embryoid somatic cells prepared in the presence of fetal bovine serum from a power quiz monkey embryonic stem cell according to the above-mentioned "Method for inducing differentiation into vascular endothelial cells and blood cells without feeder".
  • A and “specific progenitor cells” (structures consisting of sac-like structures and spherical cell populations)
  • B common to vascular endothelial progenitor cells and blood cells obtained by adhesion culture of embryoid bodies are shown.
  • the culture dish containing the “specific progenitor cells” formed in Procedure 2 above is treated with trypsin / EDTA solution (manufactured by Invitrogen Corp.), so that all cells in the culture dish are detached and collected. Then, adhesion culture was continued using differentiation medium 13 (added with cyto force-in) on a new culture dish (diameter 6 cm) coated with gelatin. Adherent cells proliferate actively And in 2 days it reached confluence. Further, after 2 days, production of floating cells became clear as shown in FIG. FIG. 20 is a phase contrast photomicrograph showing the situation in which enlarged reproduction of blood cells is being performed. Both hematopoietic stromal cells (adherent cells) and blood cells produced from them (floating cells) are confirmed.
  • cell culture was continued at a pace of twice a week while changing the medium.
  • the floating cells were collected by centrifugation and returned to the culture dish.
  • Adherent cells are detached once a week using trypsin / EDTA solution (manufactured by Invitrogen Corp.) and mixed with suspension culture in the culture supernatant. Subculture was performed while diluting 1/3.
  • Figure 21 confirms the production of cells in various stages of differentiation of myeloid cells (including myeloblasts, promyelocytes, myelocytes, retromyelocytes, neutrophils, monocytes, macrophages, etc.) .
  • CD45 antigen which is a pan-blood cell marker
  • FIG. 21F shows the expression of CD45 antigen, which is a pan-blood cell marker
  • An almost pure population consisting only of blood cells is obtained. It was confirmed.
  • CD34 positive ie, CD34 positive CD45 positive double positive cells
  • hematopoietic stem cells or hematopoietic progenitor cells equivalent to hematopoietic stem cells
  • FIG. 24 is a phase-contrast micrograph of the situation in which suspended cells and adherent cells were frozen and thawed and culture was resumed in the expanded reproduction of blood cells.
  • the presence of hematopoietic stromal cells (adherent cells) and blood cells (floating cells) produced therefrom were confirmed just as before freezing.
  • Figure 22 shows phase contrast micrographs of passable ⁇ hematopoietic stromal cells '' produced by the method described above (A), and the results of CD34 and CD45 expression examined by flow cytometry (B). Is. Both pancreatic cell marker CD45 and hematopoietic stem cell marker CD34 were almost negative and confirmed to be non-hematopoietic cells (a small amount of positive cells adhered to hematopoietic stroma cells! /, It is thought that hematopoietic stem cells are contaminated).
  • the adherent cells formed in Procedure 3 described above hardly expressed the CD45 antigen, which is a pan-blood cell marker (see FIG. 22), and were confirmed to be non-hematopoietic cells.
  • the cell morphology was heterogeneous, all of which were relatively flat, star-shaped, elongated, and contained short pseudopods and multinucleated large cells. This state was morphologically very similar to a heterogeneous cell population called “hematopoietic stroma”, which is an adherent cell obtained by culturing bone marrow blood.
  • Figure 23 shows a phase contrast micrograph of “CD34-positive and CD45-positive cells” produced by long-term culture (> 100 days) of passable “hematopoietic stem / progenitor cells” produced by the method described above.
  • True A
  • the force of mixing floating cells and adherent cells can migrate to each other, and is considered to be an equivalent cell population (ie, hematopoietic stem cells and equivalents).
  • Figure 23B shows the results of CD34 and CD45 expression on floating cells and adherent cells confirmed by flow cytometry.
  • hematopoietic stem cells are said to be “CD34-positive and CD45-positive cells”, and in hematopoietic stem cell transplantation, the cells are isolated and purified from bone marrow blood or umbilical cord blood and transplanted.
  • both floating cells and adherent cells are in the same cell lineage as hematopoietic stem cells or hematopoietic progenitor cells equivalent to hematopoietic stem cells, and hematopoietic stem cells (or hematopoietic progenitor cells equivalent to hematopoietic stem cells). ) Is not only a floating cell but also an adherent cell.
  • Differentiation medium 13 Composition: Knockout D-MEM (Knockout D-MEM (Invitrogen Corp.), 20 wt% KNOCKOUT (registered trademark) SR (Invitrogen Corp.)), O. lmM / 3 —Mercaptoethanol (manufactured by Sigma Chemical Co.), 1% non-essential amino acid solution (manufactured by Invitrogen Corp.), ImM L glutamine (manufactured by Invitrogen Corp.)) at a final concentration of 5 Ong / ml bone morphogenetic protein-4 (8 ⁇ [?
  • Example 1 After exfoliating the forceless cynomolgus embryonic stem cells maintained undifferentiated in Example 1 from the culture vessel coated with Matrigel (registered trademark) matrix in accordance with the method described in Example 6, using differentiation medium 1-4 Float overnight using a non-adhesive culture vessel (6 cm diameter culture dish, etc.) coated with poly (2-hydoxyethyl metha crylate) (Sigma) or Hydrocell (CellSeed). Cultured. The next day, the medium is changed to the differentiation medium 1–4 (without addition of cyto force in) containing the above-mentioned cytokin, and floating culture is further performed for about 2 weeks. ) Was created. The medium was changed every 3-4 during the 2-week culture period.
  • Matrigel registered trademark
  • V which is suspended in the medium, and the embryoid body or cell aggregates similar to the embryoid body) collect the culture supernatant to centrifuge the cell components, and newly prepare the differentiation medium 1
  • the suspension was suspended in 3 (no addition of site force in) and transferred to a newly prepared non-adhesive culture vessel to continue the culture.
  • the embryoid body formed in Procedure 1 above is recovered from the culture supernatant by centrifugal sedimentation, and 0.1% On a culture dish (24-well multi-well dish) coated with latin (manufactured by Sigma Chemical Co.) using differentiation medium 14 (added with cyto force in) in a CO incubator, Adhesion culture was performed at 37 ° C and 5% by volume. Thereafter, the medium was changed every 3 to 4 days.
  • the culture dish containing the “specific progenitor cells” formed in Procedure 2 above is treated with trypsin / EDTA solution (manufactured by Invitrogen Corp.), so that all cells in the culture dish are detached and collected. Then, the culture was continued on a new culture dish (diameter 6 cm) coated with gelatin using differentiation medium 1-4 (site force-in added). Adherent cells gradually grew and reached confluence after a few days. Further culturing revealed the production of floating cells.
  • Figure 25 shows a Wright-Giemsa stained image (A) and a special staining (myelin peroxidase staining (B), esterase doubled) of blood cells prepared from serum-free cynomolgus embryonic stem cells by the above method under serum-free conditions.
  • Staining (C) shows a variety of myeloid cells.
  • non-adhesive culture vessel (6 cm diameter culture dish) coated with poly (2-hydoxyethyl methacrylate) (Sigma) Etc.), or HydrocelKCellSeed) was used for suspension culture using the above-described differentiation medium (added with site force in). After 3 to 8 days, embryoid bodies (or cell aggregates similar to embryoid bodies) were formed.
  • the embryoid body (or a cell aggregate similar to embryoid body) formed in the procedure 1 was collected from the culture supernatant by centrifugation and cultured with 0.1% gelatin (manufactured by Sigma Chemical Co.). Adhesion culture was performed on a dish (culture dish having a diameter of 6 cm or 10 cm) in the CO incubator at 37 ° C and 5% by volume using the differentiation medium (site force-in added). Thereafter, the medium was changed every 3 to 4 days. Aggregates of human embryonic stem cells continue to grow while spreading on a plane, and about two weeks later, from around the center of the area where the aggregates originally existed, as shown in Fig.
  • FIG. 26 shows the production of vascular endothelial progenitor cells and “specific progenitor cells” of blood cells (structures consisting of sac-like structures and globular cell populations) from human embryonic stem cells by the method described above.
  • the sac-like structure itself is structured using a micro knife (Stem cell knife (manufactured by Swemed)). By making a small notch near the bottom of the sac-like structure that destroys the Some spherical cells were released slowly into the culture medium. When the sac-like structure is completely filled with spherical cells, the viability of the spherical cells decreases. Cultivation was continued by exchanging the medium every 3 to 4 days while collecting the spherical cells by centrifugal sedimentation of the culture supernatant.
  • Figure 27 shows the Wright-Giemsa staining image (A) of mature blood cells (floating cells) produced by the above method, and special staining (esterase double staining (B), neutrophilic alkaline phosphatase). Staining (C)).
  • A Wright-Giemsa staining image
  • B floating cells
  • C neutrophilic alkaline phosphatase
  • FIG. 28 shows the results of confirming the expression of floating cells CD34 and CD45 by flow cytometry prepared from human embryonic stem cells in the presence of fetal bovine serum by the above method. It can be seen that almost all cells express CD45, which is a pan-blood cell marker, and blood cell differentiation is induced with very high efficiency. Moreover, since about 10% of CD34 positive cells were detected, it was confirmed that hematopoietic stem cells were also present.
  • Neutrophils were concentrated from blood cells produced by the method described in Procedure 3 above by density gradient centrifugation using Lymphoprep (registered trademark) (Daiichi Chemical Co., Ltd.). When neutrophils were colored blue-violet by staining with neutrophilic alkaline phosphatase and observed under a microscope, as shown in FIG. 29, almost all of the neutrophils after concentration (B) compared to before concentration (A). Cells were confirmed to consist of neutrophils.
  • Lymphoprep registered trademark
  • FIG. 29 shows the results of concentrating neutrophils from the blood cells produced by the above method by density gradient centrifugation using Lymphoprep (registered trademark) (Daiichi Chemical Co., Ltd.).
  • Lymphoprep registered trademark
  • neutrophils are stained blue-purple by neutrophil alkaline phosphatase staining, but almost all cells are neutrophils after concentration (B) compared to before concentration (A).
  • B neutrophil alkaline phosphatase staining
  • Human embryonic stem cells consist of a differentiation medium 1 2 ⁇ Composition: Iscove's modified Dulbeccos medium (IMDM) (manufactured by Sigma Chemical Co.)], 15 wt% KNOCKOUT (registered trademark) SR [ Invitrogen (manufactured by Invitrogen Corp.), ImM ⁇ mercaptoethanol (Sigma Chemical)
  • IMDM Iscove's modified Dulbeccos medium
  • KNOCKOUT registered trademark
  • SR Invitrogen (manufactured by Invitrogen Corp.), ImM ⁇ mercaptoethanol (Sigma Chemical)
  • a non-adhesive culture vessel (6 cm diameter culture dish, etc.) coated with poly (2-hydoxyethyl methacrylate) (Sigma), Alternatively, suspension culture was performed using Hydrocell (manufactured by CellSeed) using the above-described differentiation medium (added with cyto force in). After 3 to 8 days, embryoid bodies (or cell aggregates similar to embryoid bodies) were formed.
  • the embryoid body (or a cell aggregate similar to embryoid body) formed in the procedure 1 was collected from the culture supernatant by centrifugation and cultured with 0.1% gelatin (manufactured by Sigma Chemical Co.). Adhesion culture was performed on a dish (culture dish having a diameter of 6 cm or 10 cm) in the CO incubator at 37 ° C and 5% by volume using the differentiation medium (site force-in added). Thereafter, the medium was changed every 3 to 4 days. Aggregates of human embryonic stem cells continue to grow while spreading on a flat surface, and after about two weeks, “specific progenitor cells” (capsular structures and globular cell populations) appear from around the center of the area where the aggregates originally existed. A structure consisting of
  • Fig. 30 shows the result of confirming CD45 expression in floating cells prepared from human embryonic stem cells using serum-free conditions (using KNOCKOUT (registered trademark) SR) by flow cytometry. It is. It can be seen that almost all cells express CD45, which is a pan-blood cell marker, and blood cell differentiation is induced with very high efficiency. Thus, the expression of the CD45 antigen, which is a pan-blood cell marker, was almost 100%, and in serum-free culture, an almost pure population consisting of only blood cells with very high blood cell differentiation efficiency was obtained.
  • KNOCKOUT registered trademark
  • the ectoderm component neuroepithelial cell; Fig. A, tooth enamel epithelium; Fig. D), mesoderm component (smooth muscle; Fig. B, dentin; Fig. D), endoderm component ( Since it has the three germ layers of intestinal epithelium; b, secretory gland tissue; c), it was confirmed that the tumor formed in the testis was teratoma.
  • khES-1 Human embryonic stem cells (khES-1) (3 ⁇ 10 6 cells) passaged 20 times by the undifferentiated maintenance culture method of Example 2 were transplanted into quadriceps muscles of immunodeficient mice (SICD mice). Gross tumor formation was confirmed in the quadriceps of all mice transplanted 8 weeks later. After these tumors were taken out and fixed in formalin, thin sections were prepared and subjected to hematoxylin-eosin staining (HE staining), followed by histological examination. As a result, as shown in Fig. 33, the ectoderm component (neuroepithelial cell; Fig. A, pigment epithelium; Fig. B, sebaceous gland: Fig.
  • HE staining hematoxylin-eosin stain stain staining
  • the human embryonic stem cells subcultured by the undifferentiated maintenance culture method of Example 2 have the ability to form teratoma, and that pluripotent differentiation ability is maintained! .
  • a cell agglomerate similar to the embryoid body is prepared by the hanging-drop method using the differentiation medium 1 1 (added with the site force in) using the differentiation medium of Example 4. Specifically, after collecting force quizal embryonic stem cells with a stripping solution, further react with 0.25% trypsin solution (manufactured by Invitrogen Corp.) at 37 ° C for 5 minutes. Disperse to the cellular level. Suspend 3000 force cynomolgus embryonic stem cells in 30 1 differentiation medium 1-1 (added with site force in) and use a micropipette to place it on the back of the lid of a 10 cm diameter culture dish. Pot it! /, (You can spot about 2030 spots in a single culture dish!).
  • the culture was further filled with sterilized water, and suspended in a CO incubator at 37 ° C and 5% by volume for 3 days. After 3 days, the formation of cell agglomerates can be confirmed macroscopically, so that the lid surface of the culture dish was collected and recovered, and the culture was coated with 0.1% gelatin (manufactured by Sigma Chemical Co.). Adhesion culture was performed on a dish (diameter 10 cm or 6 cm) using a differentiation medium 11 (added with cyto force in) in a CO incubator at 37 ° C and 5% by volume. Thereafter, the medium was changed every 3 to 4 days.
  • a microknife (Stem cell knif e, SweMed, etc.) to release the spherical cells in the culture medium by making a small cut near the bottom of the sac structure without destroying the structure of the sac structure itself. was released.
  • the viability of the spherical cells decreases.
  • the spherical cells were collected by sedimentation by collecting and centrifuging the culture supernatant.
  • the collected spherical cells were subjected to hematopoietic colony assembly using a colony assembly kit (Methocult (registered trademark) GF + H4535 (Stemcell Technologies In)) using a semi-solid medium containing methylcellulose.
  • a colony assembly kit Metal-Giemsa staining (A)
  • special staining myelin peroxidase staining (B)
  • Various myeloid cells were observed, ie cells at each stage of differentiation ranging from myeloblasts to mature blood cells (neutrophils and macrophages).
  • a paving stone-like cell group spread over the entire surface of the sac-like structure over time.
  • These paving stone cells and wall cells of sac-like structures are VE-cadherin, a cell indirect adhesion molecule known as a “panvascular endothelial cell marker” and a “vascular endothelial cell specific marker” as shown in the upper part of FIG. Is expressed at the intercellular junction of all cells. Note that Lampugnani et al.
  • VE-cadherin is a cell group in the “front region” where cell movement is thriving. Cell membrane localization became obscure, and a cytoplasmic expression pattern was adopted (Fig. 34, right column). On the outer side, large flat cells not expressing VE-cadherin were scattered (Fig. 34, lower right diagram, arrow). These small amounts of “non-vascular endothelial cells” are proliferating cell nu clear antigen (PCNA) -negative and no cell proliferation has occurred. In fact, over time, these VE-cadherin-negative “non-vascular endothelial cells” were quickly expelled by paving stone cells, which are VE-cadherin-positive “vascular endothelial cells”.
  • PCNA cell nu clear antigen
  • vascular endothelial cells with active proliferating ability were dominated in the culture dish within a few days. Therefore, when cells in the culture dish were collected as a lump and subculture continued, “amplification culture of vascular endothelial cells” Is possible.
  • the specific subculture procedure is as follows. Using trypsin / EDTA solution (manufactured by Invitrogen Corp.) for 5 minutes at 37 ° C, the cells were detached and recovered, and coated with 0.1% gelatin (Sigma Chemical Co.). In a new culture dish (diameter 10 cm or 6 cm), the cells were cultured using differentiation medium 11 (added with cytodynamic force in). Thereafter, the cells were subcultured every 3 to 4 days while being detached with trypsin / EDTA solution and diluted about 1/3.
  • VE-cadherin As shown in Fig. 35, it was confirmed from immunostaining that VE-cadherin was expressed at least "intracellularly" in all cells subcultured by the above procedure. Furthermore, as shown in Fig. 35, N-cadherin, an adhesion factor known to be expressed in vascular endothelial cells, is also expressed in almost all cells by immunostaining and has a clear cell membrane localization. Was shown to admit. This is a very interesting finding, as shown in Fig. 31, because the primary vascular endothelial cells obtained from the living body have been lost (see Fig. 35 and Fig. 31 for comparison).
  • VE-cadherin and PECAM1 which is a marker of mature vascular endothelial cell, express cells in S "cell membrane”. It was about 40% in the latter period and about 80% in the latter period.
  • VE-cadherin / PECAMl-positive mature vascular endothelial cells are present at an overwhelmingly higher concentration than conventional methods (2%) at any stage.
  • VE-cadherin-positive vascular endothelial cells that disappear rapidly, it is reported that amplification culture is not possible! /, In contrast to this method, VE-cadherin / PEC AMI-positive cells are passaged Concentrated with! /, It was revealed that it has distinctive features.
  • VE-cadherin As described above, all the cell populations induced to differentiate by this method express VE-cadherin at least in the cells, and there is no contamination of perisite or undifferentiated ES cells. “Mature vascular endothelial cells” that express VE-cadherin and PECAM1 in the cell membrane are produced with a purity of at least 10% in the early passage and nearly 90% in the late culture.
  • FIG. 39 and FIG. 40 show the results of sorting VE-cadherin positive cells by FACSAria (BD Biosciences) after one passage of the sac-like structure.
  • VE-cadherin positive fraction cells continue to express VE-cadherin stably in the cell membrane even after repeated passages, and after approximately 5 passages, VE-cadherin positive cells increase approximately 160 times.
  • FIG. 39B Immunostaining also confirmed the localization of VE-cadherin protein at the cell-cell junction in all cells (Fig. 39C).
  • VE-cadherin-negative cells did not express VE-cadherin on the cell membrane, but expressed VE-cadherin in the cells as described above. Furthermore, it was confirmed to be a cell population committed to vascular endothelial cells, having code-forming ability (FIG. 40B) and acetylated low-density lipoprotein uptake ability (FIG. 40C).
  • the lumen of the blood vessel is lined by immunostaining with anti-human HLA-A, B, and C monoclonal antibodies (clone W6 / 32, BioLegend), which have been confirmed to broadly cross primates. It was confirmed that vascular endothelial cells were stained (FIG. 41c). In other words, it was confirmed that the new blood vessels in the glioma tissue were formed by vascular endothelial cells derived from the force cynomolgus monkey embryonic stem cells.
  • vascular endothelial cells created from force cynomolgus monkey embryonic stem cells contribute to tumor angiogenesis in mouse transplantation experiments.
  • human embryonic stem cells (khES-3) (for one 10 cm culture dish) in an undifferentiated maintenance culture under feeder-free and site-free force-in were detached as described in Example 2. Liquid 1 Alternatively, the cells were peeled and recovered with a cell detachment solution for primate embryonic stem cells (Ribrocell). The embryo germ is obtained by performing floating culture using a general-purpose low-adsorption culture dish (one 6 cm culture dish) in the differentiation medium 11 (added with cyto force-in) using the differentiation medium of Example 4. A body-like cell aggregate was created.
  • vascular endothelial cells can be produced from human embryonic stem cells overwhelmingly more efficiently than the conventional method (2 — 7 harm IJ), and the proportion of VE-cadherin / PECAMl dual positive cells increased with passage (see Figure 42a).
  • human embryonic stem cell-derived vascular endothelial cells could be subcultured about 8 10 times as well as cynomolgus embryonic stem cell-derived vascular endothelial cells.
  • vascular endothelial cells such as code-forming ability and acetylated low density lipoprotein uptake was confirmed (see Fig. 42b).
  • the in vivo functions of the human embryonic stem cell-derived vascular endothelial cells thus prepared were evaluated.
  • a suspension of human embryonic stem cell-derived vascular endothelial cells (passage 4), several pieces of approximately 1 mm square honeycomb-like collagen sponge are repeatedly compressed and relaxed several times. Sex stem cell-derived vascular endothelial cells were filled. The cells were cultured for 2 days and then transplanted into the peritoneal cavity of an immune deficient mouse (SICD mouse) (about several pieces / mouse).
  • ISD mouse immune deficient mouse
  • FITC-labeled polymer dextran was injected from the tail vein, and a few minutes later, the collagen sponge was collected from the abdominal cavity. After formalin fixation, immunostaining was performed using anti-human HLA-A, B, C antibodies (clone W6 / 32, BioLegend) and anti-human PECAM1 antibody (Santa Cruz, sc-8306).
  • FIG. 42c when the human embryonic cell-derived vascular endothelial cells were transplanted, a “luminal structure in which the lumen was filled with FITC dextran” was confirmed in the collagen sponge.
  • This luminal structure consists of human HLA-A, B, and C antibodies that specifically recognize primates including humans without reacting with mouse cells ( Figure 42c left), and human PECAM1 antibody that is a vascular endothelial cell marker (In Fig. 42c), both stained positively. Since the human PECAM1 antibody did not stain the vascular structure of the mouse (Fig. 42c, right), this luminal structure must be a “new blood vessel composed of human embryonic stem cell-derived vascular endothelial cells” that is linked to systemic circulation. Is confirmed It is.
  • vascular endothelial cells created from human embryonic stem cells by a novel vascular endothelial cell differentiation induction method are directly involved in the formation of “functional new blood vessels linked to systemic circulation” in vivo (in new blood vessels). It was confirmed that it was incorporated).
  • Human embryonic stem cells were induced to differentiate by the method of Example 8, and cells floating in the medium were collected on day 30, and the expression of blood cell markers centered on neutrophils was analyzed by flow cytometry.
  • CD34 which is a hematopoietic stem / progenitor cell marker
  • pancreatic cell marker CD45 pancreatic cell marker CD45
  • panleukocyte marker CD33 panleukocyte marker CD33
  • neutrophil / monocyte cell marker CDl lb neutrophil / monocyte cell marker CDl lb.
  • the expression of is very high, i.e. (> 90%).
  • granulocyte markers CD66b and GPI-80 were about 60 to 80%, and expression of neutrophil specific marker CD16b was more than 30%.
  • cells positive for lactoferrin contained in the neutrophil tertiary granules were also detected.
  • hematopoietic cells prepared from human embryonic stem cells express neutrophil markers at a high rate.
  • Human embryonic stem cells were induced to differentiate by the method of Example 8, and blood cells floating in the medium on the 30th day (CD66b positive rate was about 60 to 80%) were collected.
  • About 1 ⁇ 10 6 cells were intravenously injected into NOD / SCID / ⁇ c nu11 (NOG) mice, which had previously been inflated with sterile air under the skin.
  • NOG NOG
  • zymosan A l mg / ml
  • HI-1 ⁇ 10 ng / ml
  • neutrophils prepared from human embryonic stem cells have the ability to migrate in vivo.
  • the present invention it is possible to stably provide blood cells and vascular endothelial cells suitable for blood for transfusion, transplantation materials and the like on an industrial scale. Furthermore, since the blood cells and the like of the present invention lead to the enhancement of natural healing power, the impact on the medical care and medical industry is enormous. Furthermore, if the production of safe and transfused blood that replaces the current blood donation is included, there is a possibility of expanding into the giant plant industry.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Reproductive Health (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Materials For Medical Uses (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

L'invention concerne un procédé destiné au passage d'une cellule souche embryonnaire de primate. L'invention concerne également un procédé destiné à induire la différenciation d'une cellule souche embryonnaire de primate dans une cellule endothéliale vasculaire, une cellule vasculaire ou similaire. L'invention concerne de manière spécifique un procédé comprenant les étapes consistant à : cultiver une cellule souche embryonnaire de primate dans un milieu de culture contenant un composant de type protéine, en l'absence de cellule nourricière et de cytokine, dans un récipient revêtu d'une matrice extracellulaire, afin de produire une colonie de la cellule souche embryonnaire ; libérer la colonie en présence d'un agent libérateur de cellules ; et ensemencer la colonie dans un milieu de culture qui est identique au milieu de culture utilisé à l'étape précédente. L'invention concerne également de manière spécifique un procédé comprenant les étapes consistant à : cultiver une cellule souche embryonnaire de primate dans un milieu de culture contenant ou non du sérum, en présence d'une cytokine, afin de produire un agrégat de corps embryoïdes ou de cellules semblables à des corps embryoïdes ; mener la culture adhérente de l'agrégation de corps embryoïdes ou de cellules semblables à des corps embryoïdes en présence d'une cytokine afin de produire une cellule progénitrice spécifique ; et séparer la cellule progénitrice spécifique en une cellule flottante et une cellule adhérente, produisant ainsi une cellule vasculaire ou une cellule endothéliale vasculaire.
PCT/JP2007/071811 2006-11-09 2007-11-09 Procédé destiné à la culture et au passage d'une cellule souche embryonnaire de primate, et procédé destiné à induire la différenciation de la cellule souche embryonnaire WO2008056779A1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/514,207 US20110151554A1 (en) 2006-11-09 2007-11-09 Method for culturing and subculturing primate embryonic stem cell, as well as method for inducing differentiation thereof
JP2008543145A JP5067949B2 (ja) 2006-11-09 2007-11-09 霊長類動物胚性幹細胞の培養及び継代方法、並びにその分化誘導方法
EP07831542A EP2088190A4 (fr) 2006-11-09 2007-11-09 Procédé destiné à la culture et au passage d'une cellule souche embryonnaire de primate, et procédé destiné à induire la différenciation de la cellule souche embryonnaire

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2006303929 2006-11-09
JP2006-303929 2006-11-09

Publications (1)

Publication Number Publication Date
WO2008056779A1 true WO2008056779A1 (fr) 2008-05-15

Family

ID=39364589

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2007/071811 WO2008056779A1 (fr) 2006-11-09 2007-11-09 Procédé destiné à la culture et au passage d'une cellule souche embryonnaire de primate, et procédé destiné à induire la différenciation de la cellule souche embryonnaire

Country Status (4)

Country Link
US (1) US20110151554A1 (fr)
EP (1) EP2088190A4 (fr)
JP (1) JP5067949B2 (fr)
WO (1) WO2008056779A1 (fr)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010131373A (ja) * 2008-10-30 2010-06-17 Japan Health Science Foundation 血管平滑筋細胞増殖を抑制する血管狭窄部挿入用基材
WO2010110596A2 (fr) * 2009-03-24 2010-09-30 Korea Institute Of Science And Technology Méthode de différenciation des cellules souches en cellules vasculaires et induction de l'angiogenèse grâce à cette méthode
WO2011115308A1 (fr) * 2010-03-18 2011-09-22 Kyoto University Méthode d'induction de la différenciation de cellules souches pluripotentes en cellules mésodermiques
WO2012105505A1 (fr) * 2011-01-31 2012-08-09 独立行政法人国立国際医療研究センター Cellules hépatiques hautement fonctionnelles issues de cellules souches pluripotentes, leur procédé de production et procédé de test du métabolisme/de la toxicité d'un médicament
JP2012519005A (ja) * 2009-02-27 2012-08-23 セルラー ダイナミクス インターナショナル, インコーポレイテッド 多能性細胞の分化
JP2012175962A (ja) * 2011-01-31 2012-09-13 National Institute Of Biomedical Innovation ヒト多能性幹細胞の培養方法
KR101330327B1 (ko) 2010-05-07 2013-11-14 한국생명공학연구원 줄기세포로부터 생성된 배아체를 대량 증식 및 유지하는 방법
WO2014030749A1 (fr) 2012-08-24 2014-02-27 独立行政法人理化学研究所 Procédé pour la production d'un feuillet cellulaire épithélial pigmentaire de la rétine
US9644183B2 (en) 2009-03-24 2017-05-09 Korea Institute Of Science And Technology Method for differentiation of stem cells into vascular cells and the induction of angiogenesis using the same
CN107182610A (zh) * 2017-05-25 2017-09-22 兰溪市沉默生物科技有限公司 一种提高杜鹃花成苗率栽培基质及其制备方法
JP2019083819A (ja) * 2009-12-04 2019-06-06 ステム セル アンド リジェネレイティブ メディスン インターナショナル, インコーポレイテッド ヒト胚性幹細胞由来血管芽細胞からナチュラルキラー細胞および樹状細胞を生成する方法
WO2020130068A1 (fr) * 2018-12-20 2020-06-25 住友化学株式会社 Population de cellules contenant des érythroblastes embryonnaires et son procédé de production, composition de culture cellulaire et procédé d'analyse de composé
CN113631699A (zh) * 2018-10-24 2021-11-09 赫贝细胞股份有限公司 用于生产造血谱系细胞的方法和系统

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102660495A (zh) 2006-04-14 2012-09-12 先进细胞技术公司 血管瘤集落形成细胞
US9080145B2 (en) 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
EP2185693B1 (fr) 2007-07-31 2019-07-03 Lifescan, Inc. Différenciation de cellules souches embryonnaires humaines
EP2229434B1 (fr) 2007-11-27 2011-09-07 Lifescan, Inc. Différentiation des cellules souches embryonnaires humaines
CA2959401C (fr) 2008-02-21 2021-12-07 Centocor Ortho Biotech Inc. Procedes, plaques a surface modifiee et compositions permettant la fixation, la culture et le detachement de cellules
EP3293255A1 (fr) * 2008-03-27 2018-03-14 Asterias Biotherapeutics, Inc. Différenciation de cellules souches pluripotentes de primates en cellules de lignage hématopoïétique
EP2310492B1 (fr) 2008-06-30 2015-07-22 Janssen Biotech, Inc. Différenciation de cellules souches pluripotentes
US9012218B2 (en) 2008-10-31 2015-04-21 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
CA2742268C (fr) 2008-10-31 2020-02-18 Centocor Ortho Biotech Inc. Differenciation de cellules souches embryonnaires humaines en la lignee endocrine pancreatique
RU2555538C2 (ru) 2008-11-20 2015-07-10 Сентокор Орто Байотек Инк. Культура плюрипотентных стволовых клеток на микроносителях
AU2009316583B2 (en) 2008-11-20 2016-04-21 Janssen Biotech, Inc. Methods and compositions for cell attachment and cultivation on planar substrates
BR112012001480A2 (pt) 2009-07-20 2015-09-01 Janssen Biotech Inc Diferenciação de células-tronco embriônicas humanas
US9150833B2 (en) 2009-12-23 2015-10-06 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
WO2011109279A2 (fr) 2010-03-01 2011-09-09 Centocor Ortho Biotech Inc. Procédés de purification de cellules issues de cellules souches pluripotentes
CN102884176B (zh) 2010-05-12 2017-09-05 詹森生物科技公司 人胚胎干细胞的分化
PL2611910T3 (pl) 2010-08-31 2018-06-29 Janssen Biotech, Inc Różnicowanie ludzkich embrionalnych komórek macierzystych
PL2611909T3 (pl) 2010-08-31 2018-05-30 Janssen Biotech, Inc Zróżnicowanie ludzkich embrionalnych komórek macierzystych
BR112014015417A8 (pt) 2011-12-22 2017-07-04 Janssen Biotech Inc diferenciação de células-tronco embrionárias humanas em células positivas de insulina hormonal únicas
AU2013230020B2 (en) 2012-03-07 2018-08-09 Janssen Biotech, Inc. Defined media for expansion and maintenance of pluripotent stem cells
CN104334719B (zh) 2012-06-08 2018-02-13 詹森生物科技公司 人胚胎干细胞向胰腺内分泌细胞的分化
US10487310B2 (en) 2012-06-08 2019-11-26 Riken Vessel for culturing human ES cells
JP6584956B2 (ja) 2012-12-21 2019-10-02 アステラス インスティテュート フォー リジェネレイティブ メディシン 多能性幹細胞から血小板を生産するための方法およびその組成物
RU2018116647A (ru) 2012-12-31 2018-10-24 Янссен Байотек, Инк. Культивация эмбриональных стволовых клеток человека в воздушно-жидкостной зоне взаимодействия с целью их дифференцировки в панкреатические эндокринные клетки
MX2015008619A (es) 2012-12-31 2016-01-12 Janssen Biotech Inc Suspension y agrupamiento de celulas humanas pluripotentes para la diferenciacion a celulas endocrinas pancreaticas.
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
MX2015008578A (es) 2012-12-31 2015-09-07 Janssen Biotech Inc Diferenciacion de celulas madre embrionarias humanas en celulas endocrinas pancreaticas mediante el uso de relugadores de hb9.
KR20160079072A (ko) * 2013-11-01 2016-07-05 얀센 바이오테크 인코포레이티드 췌장 내분비 세포로의 분화를 위한 인간 만능 줄기세포의 현탁 및 클러스터링
RU2694311C2 (ru) 2014-05-16 2019-07-11 Янссен Байотек, Инк. Применение малых молекул для увеличения экспрессии mafa в панкреатических эндокринных клетках
KR20170084495A (ko) * 2016-01-12 2017-07-20 건국대학교 글로컬산학협력단 줄기세포 유래 혈관세포 및 혈액모세포 비정제 동시 분화 방법
MA45479A (fr) 2016-04-14 2019-02-20 Janssen Biotech Inc Différenciation de cellules souches pluripotentes en cellules de l'endoderme de l'intestin moyen
WO2020079981A1 (fr) * 2018-10-17 2020-04-23 株式会社マンダム Procédé d'observation de glande sébacée et son utilisation
CN116688233A (zh) * 2022-02-25 2023-09-05 上海软馨生物科技有限公司 一种组织工程软骨颗粒移植物及其制备方法

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999020741A1 (fr) 1997-10-23 1999-04-29 Geron Corporation Procedes et matieres utiles pour la croissance de cellules souches primordiales de primate
WO2005065354A2 (fr) * 2003-12-31 2005-07-21 The Burnham Institute Milieu défini pour culture de cellules souches pluripotentes
WO2005085426A1 (fr) * 2004-03-04 2005-09-15 Tanabe Seiyaku Co., Ltd. Milieu en vue d'une différenciation exempte d'alimentateur et procédé de différenciation exempt d'alimentateur à partir d'une cellule souche embryonnaire de primate

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999020741A1 (fr) 1997-10-23 1999-04-29 Geron Corporation Procedes et matieres utiles pour la croissance de cellules souches primordiales de primate
WO2005065354A2 (fr) * 2003-12-31 2005-07-21 The Burnham Institute Milieu défini pour culture de cellules souches pluripotentes
WO2005085426A1 (fr) * 2004-03-04 2005-09-15 Tanabe Seiyaku Co., Ltd. Milieu en vue d'une différenciation exempte d'alimentateur et procédé de différenciation exempt d'alimentateur à partir d'une cellule souche embryonnaire de primate

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
BUZZARD, NATURAL BIOTECHNOLOGY, vol. 22, 2004, pages 381 - 382
CHADWICK ET AL., BLOOD, vol. 102, 2003, pages 906 - 915
DING V. ET AL.: "Deciphering the importance of three key media components in human embryonic stem cell cultures", BIOTECHNOL. LETT., vol. 28, April 2006 (2006-04-01), pages 491 - 495, XP019231244 *
DRAPER ET AL., NATURE BIOTECHNOLOGY, vol. 22, 2004, pages 53 - 54
JUN YAMASHITA, INFLAMMATION/REGENERATION, vol. 22, 2002, pages 509
KAUFMAN D.S. ET AL.: "Functional endothelial cells derived from rhesus monkey embryonic stem cells", BLOOD, vol. 103, 2004, pages 1325 - 1332, XP008109928 *
LAMPUGNAMI ET AL., JOURNAL OF CELL BIOLOGY, vol. 129, 1995, pages 203 - 217
LEVENBERG ET AL., PROCEEDING OF NATURAL ACADEMY OF SCIENCE, USA, vol. 99, 2002, pages 4391 - 4396
LUDWIG ET AL., NATURE BIOTECHNOLOGY, vol. 24, 2006, pages 185 - 187
NAKAHARA N. ET AL.: "Kanikuizaru Oyobi Hito ES Saibo kara no Feeder-free Baiyo ni yoru Zoketsu Saibo Bunka", SAISEI IRYO, vol. 6, no. SUPPL., 19 February 2007 (2007-02-19), pages 207 + ABSTR. NO. P-009, XP008117024 *
NAKAMURA N. ET AL.: "Kanikuizaru ES Saibo kara no Feeder-free Baiyo Jokenka ni Okeru Keidai Baiyo Kano na Kekkan Naihi Saibo eno Bunka Yudo", SAISEI IRYO, vol. 6, no. SUPPL., 19 February 2007 (2007-02-19), pages 207 + ABSTR. NO. O-11-2 P-010, XP003025829 *
REUBINOFF. B. E. ET AL.: "Embryonic stem cell lines from a human blastcysts: somatic differentiation in vitro.", NAT. BIOTECH., 2000, pages 399 - 404, XP002195338, DOI: doi:10.1038/74447
See also references of EP2088190A4
SONE ET AL., CIRCULATION, vol. 107, 2003, pages 2085 - 2088
SUEMORI, H. ET AL.: "Establishment of embryonic stem cell lines from cynomolgus monkey blastcysts produced by IVF or ICSI.", DEV. DYNAMICS, vol. 222, 2001, pages 273 - 279
TOMSON, J. A. ET AL.: "Embryonic stem cell lines derived from human blastcysts", SCIENCE, vol. 282, 1998, pages 1145 - 1147
TOMSON, J.A. ET AL.: "Isolation of a primate embryonic stem cell line.", PROC. NATL. ACAD. SCI, USA, vol. 92, 1995, pages 7844 - 7848
TOMSON, J.A. ET AL.: "Pluripotent cell lines derived from common marmoset blastcysts.", BIOLOL. REPROD., vol. 55, 1996, pages 254 - 259
UMEDA K. ET AL.: "Identification and characterization of hemoangiogenic progenitors during cynomolgus monkey embryonic stem cell differentiation", STEM CELLS, vol. 24, January 2006 (2006-01-01), pages 1348 - 1358, XP002483684 *
VOGELI ET AL., NATURE, vol. 443, 2006, pages 337 - 339
WANG L. ET AL.: "Endothelial and hematopoietic cell fate of human embryonic stem cells originates from primitive endothelium with hemangioblastic properties", IMMUNITY, vol. 21, no. 1, 2004, pages 31 - 41, XP002484358 *
YUO A.: "Reichorui ES Saibo no Feeder-free, Mukessei Baiyo o Mochiita Atarashii Mibunka Iji Zoshoku Baiyoho to Ketsueki Saibo Bunka Seigyokei no Kaihatsu", HEISEI 17 NENDO SOYAKUTO HUMAN SCIENCE KENKYU JUTEN KENKYU HOKOKUSHO, July 2006 (2006-07-01), pages 511 - 515, XP003025831 *
ZHAN X. ET AL.: "Functional antigen-presenting leucocytes derived from human embryonic stem cells in vitro", LANCET, vol. 364, 2004, pages 163 - 171, XP005070191 *
ZHANG H. ET AL.: "Efficient and repetitive production of hematopoietic and endothelial cells from feeder-free monolayer culture system of primate embryonic stem cells", BIOL. REPROD., vol. 74, February 2006 (2006-02-01), pages 295 - 306, XP008109929 *

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010131373A (ja) * 2008-10-30 2010-06-17 Japan Health Science Foundation 血管平滑筋細胞増殖を抑制する血管狭窄部挿入用基材
JP2012519005A (ja) * 2009-02-27 2012-08-23 セルラー ダイナミクス インターナショナル, インコーポレイテッド 多能性細胞の分化
WO2010110596A2 (fr) * 2009-03-24 2010-09-30 Korea Institute Of Science And Technology Méthode de différenciation des cellules souches en cellules vasculaires et induction de l'angiogenèse grâce à cette méthode
WO2010110596A3 (fr) * 2009-03-24 2011-03-10 Korea Institute Of Science And Technology Méthode de différenciation des cellules souches en cellules vasculaires et induction de l'angiogenèse grâce à cette méthode
US9644183B2 (en) 2009-03-24 2017-05-09 Korea Institute Of Science And Technology Method for differentiation of stem cells into vascular cells and the induction of angiogenesis using the same
JP7343984B2 (ja) 2009-12-04 2023-09-13 アステラス インスティテュート フォー リジェネレイティブ メディシン ヒト胚性幹細胞由来血管芽細胞からナチュラルキラー細胞および樹状細胞を生成する方法
JP2022027790A (ja) * 2009-12-04 2022-02-14 アステラス インスティテュート フォー リジェネレイティブ メディシン ヒト胚性幹細胞由来血管芽細胞からナチュラルキラー細胞および樹状細胞を生成する方法
JP2019083819A (ja) * 2009-12-04 2019-06-06 ステム セル アンド リジェネレイティブ メディスン インターナショナル, インコーポレイテッド ヒト胚性幹細胞由来血管芽細胞からナチュラルキラー細胞および樹状細胞を生成する方法
US8652845B2 (en) 2010-03-18 2014-02-18 Kyoto University Method for producing mesodermal cells by culturing under adherent conditions and without co-culture with cells from a different species in a serum-free medium
WO2011115308A1 (fr) * 2010-03-18 2011-09-22 Kyoto University Méthode d'induction de la différenciation de cellules souches pluripotentes en cellules mésodermiques
KR101330327B1 (ko) 2010-05-07 2013-11-14 한국생명공학연구원 줄기세포로부터 생성된 배아체를 대량 증식 및 유지하는 방법
JP2012175962A (ja) * 2011-01-31 2012-09-13 National Institute Of Biomedical Innovation ヒト多能性幹細胞の培養方法
WO2012105505A1 (fr) * 2011-01-31 2012-08-09 独立行政法人国立国際医療研究センター Cellules hépatiques hautement fonctionnelles issues de cellules souches pluripotentes, leur procédé de production et procédé de test du métabolisme/de la toxicité d'un médicament
WO2014030749A1 (fr) 2012-08-24 2014-02-27 独立行政法人理化学研究所 Procédé pour la production d'un feuillet cellulaire épithélial pigmentaire de la rétine
CN107182610A (zh) * 2017-05-25 2017-09-22 兰溪市沉默生物科技有限公司 一种提高杜鹃花成苗率栽培基质及其制备方法
CN113631699A (zh) * 2018-10-24 2021-11-09 赫贝细胞股份有限公司 用于生产造血谱系细胞的方法和系统
JP2022512810A (ja) * 2018-10-24 2022-02-07 ヒービセル コーポレイション 造血系統細胞を製造するための方法およびシステム
WO2020130068A1 (fr) * 2018-12-20 2020-06-25 住友化学株式会社 Population de cellules contenant des érythroblastes embryonnaires et son procédé de production, composition de culture cellulaire et procédé d'analyse de composé
JP7446242B2 (ja) 2018-12-20 2024-03-08 住友化学株式会社 胚型赤芽球を含む細胞集団及びその製造方法、細胞培養組成物並びに化合物試験法

Also Published As

Publication number Publication date
JP5067949B2 (ja) 2012-11-07
US20110151554A1 (en) 2011-06-23
EP2088190A4 (fr) 2011-01-05
EP2088190A1 (fr) 2009-08-12
JPWO2008056779A1 (ja) 2010-02-25

Similar Documents

Publication Publication Date Title
JP5067949B2 (ja) 霊長類動物胚性幹細胞の培養及び継代方法、並びにその分化誘導方法
JP6943937B2 (ja) 幹細胞培養のためのマイクロキャリア
Cheng et al. The influence of spheroid formation of human adipose-derived stem cells on chitosan films on stemness and differentiation capabilities
US20210395684A1 (en) Methods and systems for manufacturing hematopoietic lineage cells
JP4146802B2 (ja) 単球を起源に持つ、脱分化したプログラム可能な幹細胞およびそれらの製造と使用
TWI535377B (zh) Storage, culture and application of umbilical cord tissue and its derived cells
US7247477B2 (en) Methods for the in-vitro identification, isolation and differentiation of vasculogenic progenitor cells
US11261426B2 (en) Pluripotent stem cell that can be isolated from body tissue
WO2012133948A1 (fr) Composition pour thérapie cellulaire par allogreffe, ladite composition contenant une cellule souche pluripotente positive pour ssea-3 pouvant être isolée de tissu corporel
WO2021049617A1 (fr) Milieu exempt de sérum ne contenant pas d'albumine et approprié pour la culture de cellules souches hématopoïétiques humaines, et procédé de culture sans albumine
KR20100084620A (ko) 조직 재생을 위한 세포 조성물
Flippe et al. Rapid and reproducible differentiation of hematopoietic and T cell progenitors from pluripotent stem cells
Kitagawa et al. Differentiation of mesodermal cells from pluripotent stem cells
Moon et al. Differentiation of hESCs into mesodermal subtypes: vascular-, hematopoietic-and mesenchymal-lineage cells
WO2018038242A1 (fr) Méthode de culture de cellules souches pluripotentes sur une laminine spécifique
US10542743B2 (en) Isolation, expansion and characterization of wharton's jelly mesenchymal stem cells
KR100773253B1 (ko) 성체줄기세포와의 공동배양을 통한 조혈모세포의 배양 및증식방법
WO2005085426A1 (fr) Milieu en vue d'une différenciation exempte d'alimentateur et procédé de différenciation exempt d'alimentateur à partir d'une cellule souche embryonnaire de primate
JP2008141973A (ja) 心筋細胞の培養増殖法
Kelley et al. Collection and Expansion of Stem Cells
EP3564363A1 (fr) Production de mégacaryocytes dans des bioréacteurs
Espinha Bioprocess engineering of induced pluripotent stem cells for application in cell therapy and pre-clinical research
Wang Scalable expansion and erythrocyte production of human induced pluripotent stem cells
KR20090008652A (ko) 옥트(Oct)-4 발현능을 가지는 피부 유래 다분화능 성체줄기세포 및 그의 제조방법
EP2749640A1 (fr) Procédés de production de cellules ectodermiques, mésodermiques et endodermiques ainsi que de cellules souches pluripotentes, de cellules souches hématopoïétiques, de cellules de population latérale et de cellules souches mésenchymateuses et procédé permettant de dédifférencier des cellules sanguines périphériques

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07831542

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2008543145

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007831542

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12514207

Country of ref document: US